Table of Contents

 

 

 

UNITED STATES

SECURITIES AND EXCHANGE COMMISSION

Washington D.C. 20549

 

FORM 10-Q

 

(Mark One)

x       QUARTERLY REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES EXCHANGE ACT OF 1934

 

For the quarterly period ended March 31, 2017

 

OR

 

o          TRANSITION REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES EXCHANGE ACT OF 1934

 

For the transition period from             to             

 

Commission file number: 001-36294

 

uniQure N.V.

(Exact name of Registrant as specified in its charter)

 

The Netherlands

 

Not applicable

(State or other jurisdiction of incorporation or organization)

 

(I.R.S. Employer Identification No.)

 

Paasheuvelweg 25a,

1105 BP Amsterdam, The Netherlands
(Address of principal executive offices) (Zip Code)

 

+31-20-240-6000

(Registrant’s telephone number, including area code)

 

Indicate by check mark whether the registrant (1) has filed all reports required to be filed by Section 13 or 15(d) of the Securities Exchange Act of 1934 during the preceding 12 months (or for such shorter period that the registrant was required to file such reports), and (2) has been subject to such filing requirements for the past 90 days.  Yes x No o

 

Indicate by check mark whether the registrant has submitted electronically and posted on its corporate Web site, if any, every Interactive Data File required to be submitted and posted pursuant to Rule 405 of Regulation S-T (§ 232.405 of this chapter) during the preceding 12 months (or for such shorter period that the registrant was required to submit and post such files).  Yes x No o

 

Indicate by check mark whether the registrant is a large accelerated filer, an accelerated filer, a non-accelerated filer, smaller reporting company or an emerging growth company. See the definitions of “large accelerated filer” “accelerated filer” and “smaller reporting company” and “emerging growth company” in Rule 12b-2 of the Exchange Act.

 

Large accelerated filer  o

 

Accelerated filer  x

 

 

 

Non accelerated filer  o

 

Smaller reporting company  o

(do not check if smaller reporting company)

 

 

 

 

Emerging growth company  x

 

If an emerging growth company, indicate by check mark if the registrant has elected not to use the extended transition period for complying with any new or revised financial accounting standards provided pursuant to Section 13 (a) of the Exchange Act   Yes  x No  o

 

Indicate by check mark whether the registrant is a shell company (as defined in Rule 12b-2 of the Exchange Act.)   Yes o No x

 

As of May 1, 2017, the registrant had 25,475,894 shares of common shares, par value €0.05, outstanding.

 


 

 

 



Table of Contents

 

TABLE OF CONTENTS

 

 

 

Page

 

 

 

PART I — FINANCIAL INFORMATION

 

 

 

Item 1

Financial Statements

4

 

 

 

Item 2

Management’s Discussion and Analysis of Financial Condition and Results of Operations

23

 

 

 

Item 3

Quantitative and Qualitative Disclosures About Market Risk

32

 

 

 

Item 4

Controls and Procedures

32

 

 

 

PART II — OTHER INFORMATION

 

 

 

Item 1

Legal Proceedings

33

 

 

 

Item 1A

Risk Factors

33

 

 

 

Item 2

Unregistered Sales of Equity Securities and Use of Proceeds

55

 

 

 

Item 3

Defaults Upon Senior Securities

55

 

 

 

Item 4

Mine Safety Disclosures

55

 

 

 

Item 5

Other Information

55

 

 

 

Item 6

Exhibits

56

 

2



Table of Contents

 

SPECIAL CAUTIONARY NOTICE REGARDING FORWARD-LOOKING STATEMENTS

 

This Quarterly Report on Form 10-Q contains “forward-looking statements” as defined under federal securities laws. Forward-looking statements are based on our current expectations of future event and many of these statements can be identified by the use of terminology such as “believes,” “expects,” “anticipates,” “plans,” “may,” “will,” “projects,” “continues,” “estimates,” “potential,” “opportunity” and similar expressions. These forward-looking statements may be found in Part II, Item 1A “Risk Factors,” Part I, Item 2”Management’s Discussion and Analysis of Financial Condition and Results of Operations” and other sections of this Quarterly Report on Form 10-Q.

 

Forward-looking statements involve risks and uncertainties that could cause actual results to differ materially from those projected or implied. The most significant factors known to us that could materially adversely affect our business, operations, industry, financial position or future financial performance include those discussed in Part II, Item 1a”Risk Factors,” as well as those discussed in Part I, Item 2 “Management’s Discussion and Analysis of Financial Condition and Results of Operations” and elsewhere in this report, as well as other factors which may be identified from time to time in our other filings with the Securities and Exchange Commission (“SEC”) , including our most recent Annual Report on Form 10-K filed with the Securities and Exchange Commission, or SEC, on March 15, 2017, or in the documents where such forward-looking statements appear. You should carefully consider that information before you make an investment decision.

 

You should not place undue reliance on these statements, which speak only as of the date that they were made. Our actual results or experience could differ significantly from those anticipated in the forward-looking statements and from historical results, due to the risks and uncertainties described in this Quarterly Report on Form 10-Q for the quarter ended March 31, 2017 and in our Annual Report on Form 10-K for the year ended December 31, 2016, including in “ Part I, Item 1A. Risk Factors ,” as well as others that we may consider immaterial or do not anticipate at this time. These cautionary statements should be considered in connection with any written or oral forward-looking statements that we may make in the future or may file or furnish with the SEC. We do not undertake any obligation to release publicly any revisions to these forward-looking statements after completion of the filing of this Quarterly Report on Form 10-Q to reflect later events or circumstances or to reflect the occurrence of unanticipated events. All forward-looking statements attributable to us are expressly qualified in their entirety by these cautionary statements.

 

In addition, with respect to all of our forward-looking statements, we claim the protection of the safe harbor for forward-looking statements contained in the Private Securities Litigation Reform Act of 1995.

 

3



Table of Contents

 

Part I — FINANCIAL INFORMATION

 

Item 1. Financial Statements

 

uniQure N.V.

 

UNAUDITED CONDENSED CONSOLIDATED BALANCE SHEETS

 

 

 

March 31,

 

December 31,

 

 

 

2017

 

2016

 

 

 

in thousands, except share and per share amounts

 

Current assets

 

 

 

 

 

Cash and cash equivalents

 

$

120,271

 

$

132,496

 

Accounts receivable and accrued income

 

1,233

 

3,680

 

Accounts receivable and accrued income from related party

 

517

 

5,500

 

Prepaid expenses

 

1,129

 

996

 

Other current assets

 

579

 

1,274

 

Total current assets

 

123,729

 

143,946

 

Non-current assets

 

 

 

 

 

Property, plant and equipment, net

 

35,241

 

35,702

 

Intangible assets, net

 

8,357

 

8,324

 

Goodwill

 

471

 

465

 

Other non-current assets

 

1,837

 

1,828

 

Total non-current assets

 

45,906

 

46,319

 

Total assets

 

$

169,635

 

$

190,265

 

Current liabilities

 

 

 

 

 

Accounts payable

 

$

3,954

 

$

5,524

 

Accrued expenses and other current liabilities

 

7,154

 

9,766

 

Current portion of long-term debt

 

2,443

 

605

 

Current portion of deferred rent

 

697

 

684

 

Current portion of deferred revenue

 

6,225

 

6,142

 

Total current liabilities

 

20,473

 

22,721

 

Non-current liabilities

 

 

 

 

 

Long-term debt, net of current portion

 

17,920

 

19,631

 

Deferred rent, net of current portion

 

8,090

 

6,781

 

Deferred revenue, net of current portion

 

75,404

 

75,612

 

Contingent consideration

 

1,989

 

1,838

 

Other non-current liabilities

 

32

 

51

 

Total non-current liabilities

 

103,435

 

103,913

 

Total liabilities

 

123,908

 

126,634

 

Commitments and contingencies (see note 13)

 

 

 

 

 

Shareholders’ equity

 

 

 

 

 

Common shares, €0.05 par value: 60,000,000 shares authorized at March 31, 2017, and December 31, 2016, and 25,475,894 and 25,257,420 shares issued and outstanding at March 31, 2017, and December 31, 2016, respectively.

 

1,604

 

1,593

 

Additional paid-in-capital

 

466,688

 

464,653

 

Accumulated other comprehensive loss

 

(6,235

)

(6,557

)

Accumulated deficit

 

(416,330

)

(396,058

)

Total shareholders’ equity

 

45,727

 

63,631

 

Total liabilities and shareholders’ equity

 

$

169,635

 

$

190,265

 

 

The accompanying notes are an integral part of these unaudited condensed consolidated financial statements.

 

4



Table of Contents

 

uniQure N.V.

 

UNAUDITED CONDENSED CONSOLIDATED STATEMENTS OF OPERATIONS AND COMPREHENSIVE LOSS

 

 

 

Three months ended March 31,

 

 

 

2017

 

2016

 

 

 

in thousands, except share and per share amounts

 

License revenues

 

$

234

 

$

241

 

License revenues from related party

 

949

 

978

 

Collaboration revenues

 

1,580

 

1,426

 

Collaboration revenues from related party

 

558

 

1,650

 

Total revenues

 

3,321

 

4,295

 

Operating expenses:

 

 

 

 

 

Research and development expenses

 

(16,994

)

(16,706

)

Selling, general and administrative expenses

 

(6,358

)

(7,298

)

Total operating expenses

 

(23,352

)

(24,004

)

Other income

 

316

 

445

 

Loss from operations

 

(19,715

)

(19,264

)

Interest income

 

11

 

22

 

Interest expense

 

(504

)

(629

)

Foreign currency gains, net

 

(93

)

(2,188

)

Other non-operating income, net

 

29

 

317

 

Loss before income tax expense

 

(20,272

)

(21,742

)

Income tax benefit / (expense)

 

 

(557

)

Net loss

 

$

(20,272

)

$

(22,299

)

Other comprehensive loss, net of income tax:

 

 

 

 

 

Foreign currency translation adjustments net of tax impact of nil for the three months ended March 31, 2017 (three months ended March 31, 2016: $(0.6) million)

 

322

 

4,680

 

Total comprehensive loss

 

$

(19,950

)

$

(17,619

)

 

 

 

 

 

 

Basic and diluted net loss per common share

 

$

(0.80

)

$

(0.90

)

Weighted average shares used in computing basic and diluted net loss per common share

 

25,443,609

 

24,696,643

 

 

The accompanying notes are an integral part of these unaudited condensed consolidated financial statements.

 

5



Table of Contents

 

uniQure N.V.

 

UNAUDITED CONDENSED CONSOLIDATED STATEMENTS OF SHAREHOLDERS’ EQUITY

 

 

 

Common shares

 

Additional
paid-in

 

Accumulated other
comprehensive

 

Accumulated

 

Total
shareholders’

 

 

 

No. of shares

 

Amount

 

capital

 

income/(loss)

 

deficit

 

equity

 

 

 

in thousands, except share and per share amounts

 

Balance at December 31, 2016

 

25,257,420

 

$

1,593

 

$

464,653

 

$

(6,557

)

$

(396,058

)

$

63,631

 

Loss for the period

 

 

 

 

 

(20,272

)

(20,272

)

Other comprehensive income

 

 

 

 

322

 

 

322

 

Exercise of share options

 

134,974

 

7

 

434

 

 

 

441

 

Shares distributed during the period

 

83,500

 

4

 

(4

)

 

 

 

Share-based compensation expense

 

 

 

1,605

 

 

 

1,605

 

Balance at March 31, 2017

 

25,475,894

 

$

1,604

 

$

466,688

 

$

(6,235

)

$

(416,330

)

$

45,727

 

 

The accompanying notes are an integral part of these unaudited condensed consolidated financial statements.

 

6



Table of Contents

 

uniQure N.V.

 

UNAUDITED CONDENSED CONSOLIDATED STATEMENTS OF CASH FLOWS

 

 

 

Three months ended March 31,

 

 

 

2017

 

2016

 

 

 

in thousands

 

Cash flows from operating activities

 

 

 

 

 

Net loss

 

$

(20,272

)

$

(22,299

)

Adjustments to reconcile net loss to net cash used in operating activities:

 

 

 

 

 

Depreciation and amortization

 

1,794

 

1,480

 

Share-based compensation expense

 

1,605

 

2,648

 

Change in fair value of derivative financial instruments

 

105

 

(244

)

Unrealized foreign exchange results

 

174

 

2,304

 

Change in deferred taxes

 

 

557

 

Change in lease incentive

 

1,305

 

(149

)

Changes in operating assets and liabilities:

 

 

 

 

 

Accounts receivable, prepaid expenses and other current assets

 

8,154

 

449

 

Inventories

 

 

31

 

Accounts payable

 

(683

)

1,191

 

Accrued expenses and other liabilities

 

(2,584

)

(276

)

Deferred revenue

 

(1,268

)

(1,418

)

Net cash used in operating activities

 

(11,670

)

(15,726

)

Cash flows from investing activities

 

 

 

 

 

Restricted cash

 

 

(608

)

Purchase of property, plant and equipment

 

(1,969

)

(1,867

)

Net cash used in investing activities

 

(1,969

)

(2,475

)

Cash flows from financing activities

 

 

 

 

 

Proceeds from issuance of shares

 

441

 

820

 

Repayment of capital lease obligations

 

 

(48

)

Net cash generated from financing activities

 

441

 

772

 

Currency effect cash and cash equivalents

 

973

 

5,483

 

Net increase / (decrease) in cash and cash equivalents

 

(12,225

)

(11,946

)

Cash and cash equivalents at beginning of period

 

132,496

 

221,626

 

Cash and cash equivalents at end of period

 

$

120,271

 

$

209,680

 

Supplemental cash flow disclosures:

 

 

 

 

 

Cash paid for interest

 

$

413

 

$

521

 

Non-cash adjustments in purchases of intangible assets and property, plant and equipment

 

$

(949

)

$

150

 

 

The accompanying notes are an integral part of these unaudited condensed consolidated financial statements.

 

7



Table of Contents

 

1                                          General business information

 

uniQure N.V. (“uniQure” or the “Company”) was founded in 1998 by scientists at the Academic Medical Center of the University of Amsterdam. The Company is a leader in the field of gene therapy and seeks to deliver to patients suffering from rare and other devastating diseases single treatments with potentially curative results. The Company initially operated through its predecessor company, Amsterdam Molecular Therapeutics Holding N.V. (“AMT”). The Company was incorporated in January 2012 to acquire and continue the gene therapy business of AMT. Effective February 10, 2014, in connection with its initial public offering, the Company converted into a public company with limited liability and changed its legal name from uniQure B.V. to uniQure N.V. The Company is registered with the Dutch Trade Register of the Chamber of Commerce ( handelsregister van de Kamer van Koophandel en Fabrieken ) in Amsterdam, the Netherlands under number 54385229. The Company’s headquarters is in Amsterdam, the Netherlands, and its registered office is located at Paasheuvelweg 25a, Amsterdam 1105 BP, the Netherlands, and its telephone number is +31 20 240 6000.

 

Effective January 1, 2017, the Company ceased to qualify as a foreign private issuer. Since January 1, 2017, the Company files electronically with the Securities and Exchange Commission (“SEC”) its annual reports on Form 10-K, quarterly reports on Form 10-Q, current reports on Form 8-K, and any amendments to those reports filed or furnished pursuant to Section 13(a) or 15(d) of the Securities Exchange Act of 1934, as amended, which is referred to as the Exchange Act. Prior to this time, it filed its annual report on Form 20-F and furnished quarterly financial report as an exhibit on Form 6-K.

 

The Company’s common stock is listed on the NASDAQ Global Market and trades under the symbol “QURE”.

 

2                                          Summary of significant accounting policies

 

2.1                                Basis of preparation

 

The Company prepared these unaudited condensed consolidated financial statements in compliance with generally accepted accounting principles in the United States of America (“U.S. GAAP”) and applicable rules and regulations of the SEC regarding interim financial reporting. Any reference in these notes to applicable guidance is meant to refer to authoritative U.S. GAAP as found in the Accounting Standards Codification (“ASC”) and Accounting Standards Update (“ASU”) of the Financial Accounting Standards Board (“FASB”).

 

The unaudited condensed consolidated financial statements are presented in U.S. dollars, except where otherwise indicated. Transactions denominated in currencies other than U.S. dollars are presented in the transaction currency with the U.S. dollar amount included in parenthesis, converted at the foreign exchange rate as of the transaction date.

 

2.2                                Unaudited interim financial information

 

The accompanying interim condensed financial statements and related disclosures are unaudited, have been prepared on the same basis as the annual financial statements and, in the opinion of management, reflect all adjustments, which include only normal recurring adjustments, necessary for a fair statement of the financial position, results of operations and changes in financial position for the periods presented.

 

The year-end condensed balance sheet data was derived from audited financial statements, but does not include all disclosures required by U.S. GAAP. Certain information and footnote disclosures normally included in financial statements prepared in accordance with U.S. GAAP have been condensed or omitted. The condensed results of operations for the three months ended March 31, 2017, are not necessarily indicative of the results to be expected for the full year ending December 31, 2017, or for any other future year or interim period. The accompanying condensed financial statements should be read in conjunction with the audited financial statements and the related notes thereto included in the Company’s Annual Report on Form 10-K for the year ended December 31, 2016, filed with the SEC on March 15, 2017.

 

2.3                                Use of estimates

 

The preparation of the financial statements in conformity with U.S. GAAP requires management to make estimates and assumptions that affect the reported amounts of assets and liabilities, disclosure of

 

8



Table of Contents

 

contingent assets and liabilities at the date of the financial statements and reported amounts of revenues and expenses during the reporting periods. Actual results could differ from those estimates.

 

2.4                                Accounting policies

 

The principal accounting policies applied in the preparation of these unaudited condensed consolidated financial statements are described in the Company’s audited financial statements as of and for the year ended December 31, 2016, and the notes thereto, which are included in the Company’s Annual Report on Form 10-K for the year ended December 31, 2016, filed with the SEC on March 15, 2017. There have been no material changes in the Company’s significant accounting policies during the three months ended March 31, 2017.

 

2.5                                Recent accounting pronouncements

 

There have been no new accounting pronouncements or changes to accounting pronouncements during the three months ended March 31, 2017, compared to the recent accounting pronouncements described in Note 2.3.22 of the Company’s Annual Report on Form 10-K for the year ended December 31, 2016, which would be expected to materially impact the Company’s unaudited condensed consolidated financial statements.

 

3                                          Fair value measurement

 

The Company measures certain assets and liabilities at fair value, either upon initial recognition or for subsequent accounting or reporting. U.S. GAAP, requires disclosure of methodologies used in determining the reported fair values, and establishes a hierarchy of inputs used when available. The three levels of the fair value hierarchy are described below:

 

·                       Level 1 - Valuations based on unadjusted quoted prices in active markets for identical assets or liabilities that the Company has the ability to access at the measurement date.

·                       Level 2 - Valuations based on quoted prices for similar assets or liabilities in markets that are not active or models for which the inputs are observable, either directly or indirectly.

·                       Level 3 - Valuations that require inputs that reflect the Company’s own assumptions that are both significant to the fair value measurement and are unobservable.

 

To the extent that valuation is based on models or inputs that are less observable or unobservable in the market, the determination of fair value requires more judgment. Accordingly, the degree of judgment exercised by the Company in determining fair value is greatest for instruments categorized as Level 3. A financial instrument’s level within the fair value hierarchy is based on the lowest level of any input that is significant to the fair value measurement.

 

Items measured at fair value on a recurring basis include financial instruments and contingent consideration (note 3, “Fair value measurement”). The carrying amount of cash and cash equivalents, accounts receivable from collaborators, prepaid expenses, other assets, accounts payable, accrued expenses and other current liabilities reflected in the consolidated balance sheets approximate their fair values due to their short-term maturities.

 

9



Table of Contents

 

The following table sets forth the Company’s assets and liabilities that are required to be measured at fair value on a recurring basis as of March 31, 2017, and December 31, 2016:

 

 

 

Quoted prices in
active markets
(Level 1)

 

Significant other
observable inputs
(Level 2)

 

Significant
unobservable inputs
(Level 3)

 

Total

 

Classification in consolidated balance sheets

 

 

 

in thousands

 

 

 

At December 31, 2016

 

 

 

 

 

 

 

 

 

 

 

Assets:

 

 

 

 

 

 

 

 

 

 

 

Cash and cash equivalents

 

$

132,496

 

$

 

$

 

$

132,496

 

 

 

Total assets

 

132,496

 

 

 

132,496

 

 

 

Liabilities:

 

 

 

 

 

 

 

 

 

 

 

Derivative financial instruments - debt

 

 

 

11

 

11

 

Accrued expenses and other current liabilities

 

Derivative financial instruments - related party

 

 

 

51

 

51

 

Other non-current liabilities

 

Contingent consideration

 

 

 

1,838

 

1,838

 

 

 

Total liabilities

 

$

 

$

 

1,900

 

$

1,900

 

 

 

At March 31, 2017

 

 

 

 

 

 

 

 

 

 

 

Assets:

 

 

 

 

 

 

 

 

 

 

 

Cash and cash equivalents

 

$

120,271

 

$

 

$

 

$

120,271

 

 

 

Total assets

 

120,271

 

 

 

120,271

 

 

 

Liabilities:

 

 

 

 

 

 

 

 

 

 

 

Derivative financial instruments - debt

 

 

 

2

 

2

 

Accrued expenses and other current liabilities

 

Derivative financial instruments - related party

 

 

 

32

 

32

 

Other non-current liabilities

 

Contingent consideration

 

 

 

1,989

 

1,989

 

 

 

Total liabilities

 

$

 

$

 

$

2,023

 

$

2,023

 

 

 

 

Changes in Level 3 items during the three months ended March 31, 2017, and 2016, are as follows:

 

 

 

Contingent
consideration

 

Derivative financial
instruments

 

Total

 

 

 

in thousands

 

Balance at December 31, 2015

 

$

2,926

 

$

837

 

$

3,763

 

(Gains) / losses recognized in profit or loss

 

60

 

(317

)

(257

)

Currency translation effects

 

127

 

30

 

157

 

Balance at March 31, 2016

 

$

3,113

 

$

550

 

$

3,663

 

 

 

 

Contingent
consideration

 

Derivative financial
instruments

 

Total

 

 

 

in thousands

 

Balance at December 31, 2016

 

$

1,838

 

$

62

 

$

1,900

 

(Gains) / losses recognized in profit or loss

 

124

 

(29

)

95

 

Currency translation effects

 

27

 

1

 

28

 

Balance at March 31, 2017

 

$

1,989

 

$

34

 

$

2,023

 

 

Contingent consideration

 

In connection with the Company’s acquisition of InoCard GmbH in 2014, the Company recorded a contingent consideration related to amounts potentially payable to InoCard’s former shareholders. The amounts payable are contingent upon realization of the following milestones:

 

·                   Successful completion of GLP safety and toxicology study;

·                   First patient dosed in a clinical study; and

·                   Full proof-of-concept of the product in human patients after finalization of a phase I/II study.

 

10



Table of Contents

 

The valuation of the contingent liability is based on significant inputs not observable in the market such as the probability of success (“POS”) of achieving certain research milestones (estimated as probable for the first two milestones as of the balance sheet date), the time at which the research milestones are expected to be achieved (ranging from 2018 to 2021), as well as the 30% discount rate applied, which represents a Level 3 measurement. Varying, next to the passing of time, the unobservable inputs results in the following fair value changes:

 

 

 

March 31,

 

December 31,

 

 

 

2017

 

2016

 

 

 

in thousands

 

Change in fair value

 

 

 

 

 

Moving out of all milestones by 6 months

 

$

(226

)

$

(209

)

Increasing the POS for the first milestone by 20%

 

398

 

367

 

Decreasing the POS for the first milestone by 20%

 

(398

)

(367

)

Reducing the discount rate from 30% to 20%

 

636

 

638

 

Increasing the discount rate from 30% to 40%

 

(305

)

(309

)

 

Derivative financial instruments

 

The Company issued derivative financial instruments related to its collaboration with Bristol-Meyers Squibb Company (“BMS”) and in relation to the issuance of the Hercules Technology Growth Corp. (“Hercules”) loan facility. The fair value of these derivative financial instruments as of March 31, 2017, was $0.0 million (December 31, 2016: $0.1 million), both of which are described in more details below.

 

There were no significant changes in (un)observable inputs, nor in the sensitivity of the fair value from (un)observable inputs as at March 31, 2017, compared to December 31, 2016.

 

BMS collaboration

 

On April 6, 2015, the Company entered into a number of agreements with BMS (the “BMS Agreements”). Pursuant to the terms of the BMS Agreements the Company granted BMS two warrants:

 

·                   A warrant allowing BMS to purchase a specific number of uniQure common shares such that its ownership will equal 14.9% immediately after such purchase. The warrant can be exercised on the later of (i) the date on which the Company receives from BMS the Target Designation Fees (as defined in the collaboration agreements) associated with the first six New Targets (as defined in the collaboration agreements); and (ii) the date on which BMS designates the sixth New Target.

·                   A warrant allowing BMS to purchase a specific number of uniQure common shares such that its ownership will equal 19.9% immediately after such purchase. The warrant can be exercised on the later of (i) the date on which uniQure receives from BMS the Target Designation Fees associated with the first nine New Targets; and (ii) the date on which BMS designates the ninth New Target.

 

Pursuant to the terms of the BMS Agreements the exercise price, in respect of each warrant, is equal to the greater of (i) the product of (A) $33.84, multiplied by (B) a compounded annual growth rate of 10% and (ii) the product of (A) 1.10 multiplied by (B) the VWAP for the 20 trading days ending on the date that is five trading days prior to the date of a notice of exercise delivered by BMS.

 

Hercules loan facility

 

On June 14, 2013, the Company entered into a venture debt loan facility with (the “Original Facility”) with Hercules Technology Growth Capital, Inc. (“Hercules”) pursuant to a Loan and Security Agreement (the “Loan Agreement”) which included a warrant. The warrant was not closely related to the host contract and was accounted for separately as a derivative financial liability measured at fair value though profit or loss. The warrant included in the Original Facility remained in place following the 2014 and 2016 amendments of the loan.

 

11



Table of Contents

 

4                                          Collaboration arrangements and concentration of credit risk

 

The Company generates all of its collaboration and license revenues from its Collaboration and License Agreement with BMS, its Co-Development Agreement for AMT-060 with Chiesi Farmaceutici S.p.A. (“Chiesi”) and its Glybera Commercialization Agreement with Chiesi. The Company agreed to terminate the Glybera Commercialization Agreement. See note 14 for additional information regarding the termination of this agreement.

 

Since June 2015, BMS has been considered a related party given the significance of its equity investment in the Company (exceeding 5%).

 

Services to the Company’s two collaboration partners are rendered by the Dutch operating entity. Total collaboration and license revenue generated from these partners are as follows:

 

 

 

Three months ended March 31,

 

 

 

2017

 

2016

 

 

 

in thousands

 

Bristol Myers Squibb

 

$

1,507

 

$

2,628

 

Chiesi Farmaceutici S.p.A

 

1,814

 

1,667

 

Total

 

$

3,321

 

$

4,295

 

 

Amounts owed by these partners in relation to the collaboration are as follows:

 

 

 

March 31,

 

December 31,

 

 

 

2017

 

2016

 

 

 

in thousands

 

Bristol Myers Squibb

 

$

517

 

$

5,500

 

Chiesi Farmaceutici S.p.A

 

1,233

 

3,680

 

Total

 

$

1,750

 

$

9,180

 

 

BMS collaboration

 

In May 2015, the Company closed a Collaboration and License Agreement with BMS (the “BMS Collaboration Agreement”) that provides exclusive access to the Company’s gene therapy technology platform for multiple targets in cardiovascular (and other) diseases. The collaboration included the Company’s proprietary gene therapy program for congestive heart failure which aims to restore the heart’s ability to synthesize S100A1, a calcium sensor and master regulator of heart function, and thereby improve clinical outcomes for patients with reduced ejection fraction. Beyond cardiovascular diseases, the agreement also included the potential for a target exclusive collaboration in other disease areas. In total, the companies may collaborate on ten targets, including S100A1.

 

The Company is leading the discovery, non-clinical, analytical and process development effort and is responsible for manufacturing of clinical and commercial supplies using the Company’s vector technologies and industrial, proprietary insect-cell based manufacturing platform, while BMS leads the clinical development and regulatory activities across all programs and reimburses the Company for all research and development efforts. BMS will be solely responsible for commercialization of all products from the collaboration.

 

12



Table of Contents

 

The Company evaluated the BMS Collaboration Agreement and determined that it is a revenue arrangement with multiple elements. The Company’s substantive deliverables under the BMS Collaboration Agreement include an exclusive license to its technology in the field of cardiovascular disease, research and development services for specific targets chosen by BMS and general development of the Company’s proprietary vector technology, participation in the Joint Steering Committee, and clinical and commercial manufacturing. The Company concluded that the BMS Collaboration Agreement consists of three units of accounting, including (i) technology (license and target selections), know-how and manufacturing in the field of gene therapy and development and active contribution to the development through the Joint Steering Committee participations, (ii) provision of employees, goods and services for research activities for specific targets and (iii) clinical and commercial manufacturing. The Company determined that the license does not have stand-alone value to BMS without the Company’s know-how and manufacturing technology through the participation of the Joint Steering Committee and accordingly, they were combined into one unit of accounting.

 

License revenue — BMS

 

As of May 21, 2015, the effective date of the BMS Collaboration Agreement, the Company recorded deferred revenue of $60.1 million. On July 31, 2015, BMS selected the second, third and fourth collaboration targets, triggering a $15.0 million target designation payment to the Company. The Company is entitled to $16.5 million in aggregate of target designation payments upon selection of the fifth through tenth collaboration target. The Company will also be eligible to receive research, development and regulatory milestone payments of up to $254.0 million for the first target and up to $217.0 million for the other selected targets if and when achieved. The Company determined that the contingent payments under the BMS Collaboration Agreement relating to research, development and regulatory milestones do not constitute substantive milestones and will not be accounted for under the milestone method of revenue recognition. The events leading to these payments solely depend on BMS’ performance. Accordingly, any revenue from these contingent payments would be allocated to the first unit of accounting noted above and recognized over the expected performance period.

 

License revenue is recognized over an expected performance period of 19 years on a straight-line basis commencing on May 21, 2015. The expected performance period is reviewed quarterly and adjusted to account for changes, if any, in the Company´s estimated performance period. The estimated performance period did not change in the three months ended March 31, 2017.

 

The Company recognized $0.9 million license revenue for the three months ended March 31, 2017, and $1.0 million in the same period 2016.

 

Additionally, the Company is eligible to receive net sales-based milestone payments and tiered high single to low double-digit royalties on product sales. These revenues will be recognized when earned. The royalty term is determined on a licensed-product-by-licensed-product and country-by-country basis and begins on the first commercial sale of a licensed product in a country and ends on the expiration of the last to expire of specified patents or regulatory exclusivity covering such licensed product in such country or, with a customary royalty reduction, ten years after such first commercial sale if there is no such exclusivity.

 

Collaboration revenue — BMS

 

The Company provides target-specific research and development services to BMS. Collaboration revenue related to these contracted services is recognized when earned.

 

The Company generated $0.6 million collaboration revenue for the three months ended March 31, 2017, and $1.7 million in the same period 2016.

 

Manufacturing revenue — BMS

 

BMS and the Company also entered into a term sheet for the Company to supply gene therapy products during the clinical and commercial phase to BMS. Revenues from product sales will be recognized when earned. To date the Company has not supplied any commercial product to BMS.

 

13



Table of Contents

 

Chiesi collaboration

 

In 2013, the Company entered into two agreements with Chiesi, a family-owned Italian pharmaceutical company, one for the co-development and commercialization of the AMT-060 program (the “Collaboration Agreement”) and one for the commercialization of Glybera (the “Glybera Agreement”, and together with the Collaboration Agreement, the “Chiesi Agreements”). The Company has retained full rights in the United States, Canada and Japan under the Chiesi Agreements. The Company evaluated the Chiesi Agreements and determined that they are a revenue arrangement with multiple elements. The Company’s substantive deliverables under the Chiesi Agreements include an exclusive license to its technology, research and development services, and commercial manufacturing. The Company concluded that the Chiesi Agreements consists of three units of accounting, including (i) co-development and active contribution to the collaboration by providing technology access and know-how in the field of gene therapy, (ii) provision of employees, goods and services for research and development activities and (iii) commercial manufacturing.

 

License revenue — Chiesi

 

Upon closing of the Chiesi Agreements on June 30, 2013, the Company received €17.0 million ($22.1 million) in non-refundable up-front payments. The Company determined that the up-front payments received from Chiesi constituted a single unit of accounting. The up-front payments are amortized on a straight-line basis and presented as license revenue over a period from July 2013, through September 2032, the date of expiration of the last intellectual property protection related to the Company’s manufacturing process at such date.

 

The Company recognized $0.2 million license revenue for the three months ended March 31, 2017, and $0.2 million in the same period 2016.

 

Collaboration revenue — Chiesi

 

Chiesi reimburses the Company for 50% of the agreed research and development efforts related to AMT-060, which is presented as collaboration revenue. Once regulatory approval has been obtained, Chiesi will distribute AMT-060 and the Company is entitled to receive a fixed mid double digit royalty as a percentage of Chiesi sales. The Company estimates that the amount it would retain, net of manufacturing costs, third-party royalties and related amounts, will be between 25% and 35% of the revenues from sales realized by Chiesi, varying by country of sale.

 

The Company generated $1.6 million collaboration revenue for the three months ended March 31, 2017, and $1.4 million in the same period 2016 from the co-development of AMT-060.

 

Manufacturing revenue — Chiesi

 

Pursuant to the Glybera Agreement, the Company to supply Glybera to Chiesi. Revenues from product sales, presented as collaboration revenue, will be recognized when earned. In the three months ended March 31, 2017, the Company recognized no revenue from product sales to Chiesi and nil in the same period 2016.

 

14



Table of Contents

 

5                                          Property, plant and equipment

 

The following table presents the Company’s property, plant and equipment as of March 31, 2017, and December 31, 2016:

 

 

 

March 31,

 

December 31,

 

 

 

2017

 

2016

 

 

 

in thousands

 

Leasehold improvements

 

$

31,286

 

$

30,582

 

Laboratory equipment

 

14,817

 

14,166

 

Office equipment

 

2,733

 

2,710

 

Construction-in-progress

 

140

 

313

 

Total property, plant, and equipment

 

48,976

 

47,771

 

Less accumulated depreciation

 

(13,735

)

(12,069

)

Property, plant and equipment, net

 

$

35,241

 

$

35,702

 

 

Total depreciation expense of $1.7 million for the three months ended March 31, 2017, and $1.4 million in the same period 2016 was charged to research and development expense to the extent it relates to the Company’s manufacturing facility and equipment, and the remainder was charged to selling, general and administrative expense.

 

6                                          Intangible assets

 

The Company’s intangible assets include acquired licenses and acquired research and development (“acquired R&D”) and are presented in the following table:

 

 

 

March 31,

 

December 31,

 

 

 

2017

 

2016

 

 

 

in thousands

 

Licenses

 

$

7,910

 

$

7,799

 

Acquired research & development

 

4,977

 

4,908

 

Total intangible assets

 

12,887

 

12,707

 

Less accumulated amortization

 

(4,530

)

(4,383

)

Intangible assets, net

 

$

8,357

 

$

8,324

 

 

The amortization expense of $0.1 million for the three months ended March 31, 2017, and $0.1 million in the same period 2016 is included in research and development expenses.

 

7                                          Accrued expenses and other current liabilities

 

Accrued expenses and other current liabilities include the following items:

 

 

 

March 31,

 

December 31,

 

 

 

2017

 

2016

 

 

 

in thousands

 

Accruals for services provided by vendors-not yet billed

 

$

3,775

 

$

4,150

 

Personnel related accruals

 

2,247

 

4,381

 

Social security and other taxes

 

1,102

 

1,178

 

Other current liabilities

 

30

 

57

 

Total

 

$

7,154

 

$

9,766

 

 

15



Table of Contents

 

In November 2016, the Company announced a plan to restructure its activities as a result of a company-wide strategic review with the aim of refocusing its pipeline, consolidating its manufacturing capabilities into its Lexington, Massachusetts site, reducing operating costs and enhancing overall execution. Following the announcement of the plan, the Company recognized an accrual for termination benefits contractually agreed with four executives of $1.1 million. In January 2017, the Company entered into termination agreements with certain non-executive employees and recognized related termination costs of $0.5 million for services rendered by these employees during 2017. The change in the accrual of termination benefits (recognized within research and development expenses) for the three months ended March 31, 2017 was:

 

 

 

Accrued
termination
benefits

 

 

 

in thousands

 

Balance at December 31, 2016

 

$

1,148

 

Accrued through profit and loss

 

241

 

Payments

 

(977

)

Currency translation effects

 

6

 

Balance at March 31, 2017

 

$

418

 

 

8                                          Long-term debt

 

On June 14, 2013, the Company entered into a venture debt loan facility with Hercules, which was amended and restated on June 26, 2014, and again on May 6, 2016 (“2016 Amended Facility”). The 2016 Amended Facility increased the total potential commitment from Hercules from $20.0 million to up to $40.0 million, of which $20.0 million was outstanding as of March 31, 2017, and December 31, 2016, and extended the maturity date from June 30, 2018, to May 1, 2020. The Company has not drawn down any additional amounts as of March 31, 2017, following the May 2016 amendment. The interest rate is adjustable and is the greater of (i) 8.25% or (ii) 8.25% plus the prime rate less 5.25%. Under the 2016 Amended Facility, the interest rate will initially be 8.25% per annum with a back-end fee of 4.85% and facility fee of 0.75% of the outstanding loan amounts. The interest-only payment period is set at November 2017, but can be extended to May 2018 upon the Company raising a cumulative $30.0 million in up-front corporate payments and/or proceeds from equity financings (“Raisings”), and further extended to November 2018 upon the Company raising a cumulative $50.0 million from the Raisings.

 

The amortized cost of the 2016 Amended Loan as of March 31, 2017, was $20.4 million and $20.2 million as of December 31, 2016, and is recorded net of discount and debt issuance costs. The foreign currency gain on the loan in the three months ended March 31, 2017, was $0.3 million and a gain of $0.9 million in the same period 2016. The fair value of the loan approximates its carrying amount, given the impact of discounting is insignificant as the loan is amortized at a market conforming interest rate.

 

In the three months ended March 31, 2017, an amount of $0.5 million and $0.6 million in the same period 2016 was recorded as interest expense in relation to the 2016 Amended Facility.

 

As a covenant in the 2016 Amended Facility, the Company has periodic reporting requirements and is required to keep a minimum cash balance deposited in bank accounts in the United States, equivalent to the lesser of the outstanding balance of principal due and 50% of worldwide cash reserves. This restriction on the cash reserves only relates to the location of the cash reserves, and such cash reserves can be used at the discretion of the Company. In combination with other convents, the 2016 Amended Facility restricts the Company’s ability to, among other things, incur future indebtedness and obtain additional debt financing, to make investments in securities or in other companies, to transfer assets, to perform certain corporate changes, to make loans to employees, officers and directors, and to make dividend payments and other distributions. The Company secured the facilities by pledging the shares in its subsidiaries, substantially all its receivables, moveable assets as well as the equipment, fixtures, inventory and cash of uniQure Inc.

 

16



Table of Contents

 

9                                          Share-based compensation

 

The Company recognized share-based compensation expense totaling $1.6 million and $2.6 million during the three months ended March 31, 2017, and 2016, respectively.

 

Share-based compensation expense recognized by classification included in the consolidated statements of operations and comprehensive loss was as follows:

 

 

 

Three months ended March 31,

 

 

 

2017

 

2016

 

 

 

in thousands

 

Research and development - employees

 

$

687

 

$

859

 

Selling, general and administrative - employees

 

918

 

1,119

 

Research and development - non-employees

 

 

670

 

Total

 

$

1,605

 

$

2,648

 

 

Share-based compensation expense recognized by award type was as follows:

 

 

 

Three months ended March 31,

 

 

 

2017

 

2016

 

 

 

in thousands

 

Award type

 

 

 

 

 

Share options

 

$

826

 

$

2,384

 

Restricted share units (“RSUs”)

 

516

 

152

 

Performance share units (“PSUs”)

 

263

 

112

 

Total

 

$

1,605

 

$

2,648

 

 

As of March 31, 2017, the unrecognized compensation costs related to unvested awards under the various share-based compensation plans were:

 

 

 

Unrecognized
compensation costs

 

Weighted-average
remaining period
for recognition (in
years)

 

 

 

in thousands

 

 

 

Award type

 

 

 

 

 

Stock options

 

$

8,324

 

2.85

 

Restricted stock units

 

3,369

 

2.22

 

Performance stock units

 

2,703

 

2.60

 

Total

 

$

14,396

 

2.66

 

 

The Company satisfies the exercise of share options and vesting of RSUs and PSUs through newly issued shares.

 

The Company’s share-based compensation plans include the 2014 Amended and Restated Share Option Plan (the “2014 Plan”) and inducement grants under Rule 5653(c)(4) of the NASDAQ Global Market with characteristics similar to the 2014 Plan (classified as “Other Plans”). The Company previously had a 2012 Equity Incentive Plan (the “2012 Plan”) and issued options to purchase common shares to the shareholders of 4D in connection with a collaboration and license agreement between the Company and 4D dated as of January 2014 (classified as Other Plans).

 

17



Table of Contents

 

2012 Plan

 

The following table summarizes option activity under the Company’s 2012 Plan for the three months ended March 31, 2017:

 

 

 

2012 Plan

 

 

 

Options

 

Weighted average
exercise price

 

 

 

 

 

 

 

Outstanding at December 31, 2016

 

483,006

 

5.13

 

Exercised

 

(134,974

)

3.07

 

Outstanding, fully vested and exercisable at March 31, 2017

 

348,032

 

5.92

 

 

Options exercised during the three months ended March 31, 2017, resulted in total proceeds to the Company of $0.4 million.

 

2014 Plan

 

Share options

 

The following table summarizes option activity under the Company’s 2014 Plan for the three months ended March 31, 2017:

 

 

 

2014 plan

 

 

 

Options

 

Weighted average
exercise price

 

 

 

 

 

 

 

Outstanding at December 31, 2016

 

1,812,766

 

$

12.47

 

Granted

 

508,500

 

$

5.63

 

Forfeited

 

(40,371

)

$

12.76

 

Expired

 

(12,952

)

$

18.30

 

Outstanding at March 31, 2017

 

2,267,943

 

$

10.90

 

Fully vested and exercisable

 

746,557

 

$

13.01

 

Outstanding and expected to vest

 

1,521,386

 

$

9.86

 

 

During the three months ended March 31, 2017, the Company granted options to purchase an aggregate of 508,500 common shares with an aggregate weighted average grant date fair value of $1.6 million. This included an option to purchase 175,000 options common shares the Company granted to its Chief Executive Officer and options to purchase an aggregate 69,000 common shares that the Company granted to its non-executive directors, each such option vesting after one year from the date of grant.

 

The fair value of each option issued was estimated at the date of grant using the Hull & White option pricing model with the following weighted-average assumptions:

 

 

 

Three months ended March 31,

 

 

 

2017

 

2016

 

Assumptions

 

 

 

 

 

Expected volatility

 

75%

 

75%

 

Expected terms (in years)

 

10 years

 

10 years

 

Risk free interest rate

 

2.53% - 2.70%

 

0.16% - 0.54%

 

Expected dividends

 

0%

 

0%

 

 

18



Table of Contents

 

Restricted Share Units (RSUs)

 

The following table summarizes the RSUs activity for the three months ended March 31, 2017:

 

 

 

Number of shares

 

Weighted average
grant-date fair
value

 

Undistributed at December 31, 2016

 

307,063

 

$

9.11

 

Granted

 

285,500

 

$

5.95

 

Distributed

 

(25,000

)

$

18.21

 

Forfeited

 

(375

)

$

13.03

 

Undistributed at March 31, 2017

 

567,188

 

$

7.12

 

 

During the three months ended March 31, 2017, the Company granted an aggregate of 285,500 RSUs with an aggregate weighted average grant date fair value of $1.7 million. The grants include 69,000 RSUs granted to non-executive directors, each such grant to vest after one year, and 175,000 RSUs granted to the Company’s Chief Executive Officer, which vest in equal tranches on February 21, 2018, and February 21, 2019.

 

During the three months ended March 31, 2017, the Company distributed 25,000 common shares in connection with the settlement of vested RSUs.

 

Performance Share Units (PSUs)

 

The following table summarizes the PSUs activity for the three months ended March 31, 2017:

 

 

 

Number of shares

 

Weighted average
grant-date fair
value

 

Undistributed at December 31, 2016

 

111,564

 

$

5.76

 

Distributed

 

(58,500

)

$

5.76

 

Undistributed at March 31, 2017

 

53,064

 

$

5.76

 

 

In January 2017, the Company awarded an aggregate of 423,500 PSUs to members of its senior management, including 162,500 PSUs to its Chief Executive Officer. As awarding these PSUs is discretionary based on the Board’s assessment of 2017 performance, these PSUs are not recorded in the above table.

 

In September 2016, the Company awarded 61,560 PSUs to its Chief Executive Officer, subject to the successful implementation of the strategic plan. As awarding thee PSUs is discretionary based on the Board’s assessment of the Chief Executive Officer’s 2017 performance, these PSUs are also not recorded in the above table.

 

19



Table of Contents

 

Other Plans

 

The following table summarizes option activity under the Company’s Other Plans for the three months March 31, 2017:

 

 

 

Other plans

 

 

 

Options

 

Weighted average
exercise price

 

 

 

 

 

 

 

Outstanding at December 31, 2016

 

187,500

 

$

17.93

 

Granted

 

150,000

 

$

5.31

 

Outstanding at March 31, 2017

 

337,500

 

$

12.32

 

Fully vested and exercisable

 

62,500

 

$

27.82

 

Outstanding and expected to vest

 

275,000

 

$

8.80

 

 

Under Rule 5653(c)(4) of the NASDAQ Global Market, the Company grants share options to certain employees as a material inducement to enter into employment with the Company. During the three months ended March 31, 2017, the Company granted 150,000 options with an aggregate weighted average grant date fair value of $0.5 million. No options were exercised during the three months ended March 31, 2017.

 

The fair value of the inducement grant options was estimated at the date of grant using the Hull & White option pricing model with the same assumptions as used in determining the fair value of options issued under the 2014 Plan.

 

10                                   Income taxes

 

Deferred tax assets and deferred tax liabilities are recognized based on the expected future tax consequences temporary differences between the financial statement carrying amounts and the income tax basis of assets and liabilities, using current statutory rates. A valuation allowance is recorded against deferred tax assets if it is more likely than not that some or all of the deferred tax assets will not be realized. Due to the uncertainty surrounding the realization of the favorable tax attributes in future tax returns, the Company has recorded a full valuation allowance against the Company’s otherwise recognizable net deferred tax assets.

 

11                                   Basic and diluted earnings per share

 

Diluted earnings per share is calculated by adjusting the weighted average number of common shares outstanding, assuming conversion of all potentially dilutive common shares. As the Company has incurred a loss, all potentially dilutive common shares would have an antidilutive effect, if converted, and thus have been excluded from the computation of loss per share.

 

 

 

March 31,

 

March 31,

 

 

 

2017

 

2016

 

 

 

common shares

 

BMS warrants

 

3,587,333

 

3,442,655

 

Warrants

 

37,175

 

37,175

 

Stock options under 2012 Plan

 

348,032

 

964,155

 

Stock options under 2014 Plan

 

2,267,943

 

1,567,236

 

Stock options (other)

 

337,500

 

1,000,000

 

RSUs and PSUs

 

620,252

 

195,940

 

Total potential dilutive common shares

 

7,198,235

 

7,207,161

 

 

20



Table of Contents

 

12                                   Leases

 

The Company leases various office space and laboratory space under operating lease agreements, expiring at various dates through 2032. These leases are described in more detail below.

 

Rent expense for the three months ended March 31, 2017, was $1.0 million and $0.8 million in the same period 2016. Rent expense is calculated on a straight-line basis over the term of the leases, and takes into account $11.8 million of lease incentives received.

 

Aggregate minimum lease payments for the calendar years ending December 31, 2017, 2018, 2019, 2020 and 2021 and beyond at March 31, 2017 were as follows:

 

 

 

in thousands

 

2017 (nine months remaining)

 

$

1,746

 

2018

 

4,132

 

2019

 

3,675

 

2020

 

3,729

 

2021 and beyond

 

26,400

 

Total minimum lease payments

 

$

39,682

 

 

Lexington, Massachusetts

 

In July 2013, uniQure entered into a lease for a facility in Lexington, Massachusetts, United States. The term of the lease commenced in November 2013 and was set for 10 years and is non-cancellable. The lease for this facility terminates in 2024, and subject to the provisions of the lease, may be renewed for two subsequent five-year terms. The Company expects to complete the qualification of its approximately 53,000 square feet manufacturing facility in 2017. The future aggregate minimum lease payments under the non-cancellable term of the lease amount to $14.0 million. As of March 31, 2017, the Company recorded a total deferred rent of $6.1 million (December 31, 2016: $6.2 million), with a current element of $0.7 million (December 31, 2016: $0.7 million) related to lease incentives received. The lease provides for annual minimum increases in rent, based on a consumer price index.

 

Paasheuvelweg, Amsterdam

 

In March 2016, the Company entered into a 16-year lease for a facility in Amsterdam, the Netherlands, and amended this agreement in June 2016. The Company expects to consolidate its three Amsterdam sites into the new site by the end of May 2017. The lease for this facility terminates in 2032, with an option to extend for further periods of five years. Following the completion of its restructuring by the end of 2017, the Company will seek to sublease parts of the facility. The future aggregate minimum lease payments under the non-cancellable term of the lease amount to $24.8 million. The lease contract provides for annual minimum increases in rent, based on a consumer price index.

 

Meibergdreef, Amsterdam

 

In April 2015, uniQure entered into a lease with Jan Snel B.V. for a laboratory facility of approximately 9,300 square feet, located at the Meibergdreef in Amsterdam, the Netherlands. The minimum lease period terminates in September 2018. The future aggregate minimum lease payments under the non-cancellable term of the lease amount to $0.9 million.

 

13                                   Other commitments

 

The Company’s predecessor entity received a technical development loan from the Dutch government in relation to the development of Glybera. The Company is required to repay the grant through a percentage of revenue derived from product sales of Glybera up to December 31, 2019. Any grant balance remaining at this date will be forgiven. The Company decided not to renew its marketing authorization for Glybera in the European Union, which expires on October 25, 2017. The Company does not expect to derive any revenue from Glybera after that time.

 

21



Table of Contents

 

14                                   Subsequent event

 

On April 20, 2017, the Company announced that it will not pursue the renewal of the Glybera® (alipogene tiparvovec) marketing authorization in Europe when it is scheduled to expire on October 25, 2017. The Company will continue to supply product to Chiesi to treat any patients that are approved for treatment prior to October 25, 2017, and will be responsible for terminating the Phase IV post-approval study. According to the termination agreement executed in April 2017, the Company will be required to make installment payments to Chiesi totaling $2.2 million through January 2019. Additionally, the Company may owe up to an additional $1.8 million depending on the number of patients treated with Glybera prior to its withdrawal, if any. Any such payments related to additional patients treated will be payable no earlier than April 1, 2019, in accordance with a schedule to be agreed upon by the parties.

 

22



Table of Contents

 

Item 2. Management’s Discussion and Analysis of Financial Condition and Results of Operations

 

The following Management’s Discussion and Analysis of Financial Condition and Results of Operations (“MD&A”) is intended to help the reader understand our results of operations and financial condition. This MD&A is provided as a supplement to, and should be read in conjunction with, our unaudited condensed consolidated financial statements and the accompanying notes thereto and other disclosures included in this Quarterly Report on Form 10-Q, including the disclosures under Part II, Item 1A “Risk Factors”, and our audited financial information and the notes thereto included in our Annual Report on Form 10-K for the year ended December 31, 2016, which was filed with the Securities and Exchange Commission, or the SEC, on March 15, 2017. Our unaudited condensed consolidated financial statements have been prepared in accordance with generally accepted accounting principles in the US (“U.S. GAAP”) and unless otherwise indicated are presented in U.S. dollars.

 

Overview

 

We are a leader in the field of gene therapy, seeking to develop single treatments with potentially curative results for patients suffering from genetic and other devastating diseases. We are advancing a focused pipeline of innovative gene therapies that have been developed both internally and through partnerships, such as our collaboration with Bristol Myers-Squibb focused on cardiovascular diseases. We have established clinical proof-of-concept in our lead indication, hemophilia B, and achieved preclinical proof-of-concept in Huntington’s disease. We believe our validated technology platform and manufacturing capabilities provide us distinct competitive advantages, including the potential to reduce development risk, cost and time to market. We produce our AAV-based gene therapies in our own facilities with a proprietary, commercial-scale, consistent, manufacturing process. We believe our Lexington, Massachusetts-based facility is one of the world’s leading, most versatile, gene therapy manufacturing facilities.

 

Business Developments

 

Below is a summary of our significant business developments in the three months ended March 31, 2017:

 

·                   On January 25, 2017, we appointed Dr. Alexander Kuta as our Senior Vice President of regulatory affairs.

·                   On January 30, 2017, we received breakthrough therapy designation from the Food and Drug Administration (“FDA”) for our AMT-060 program in hemophilia B. The designation was based on results from the ongoing, dose-ranging Phase I/II study that show sustained increases in Factor IX (FIX), reductions in FIX replacement usage and a near cessation of spontaneous bleeding in patients with severe disease at up to 12 months follow-up. During the first quarter of 2017, the Company initiated discussions with the FDA regarding the data from its ongoing Phase I/II study and the design of a potential pivotal study.

·                   On March 3, 2017, our Lexington, Massachusetts gene therapy manufacturing facility was recognized by Frost & Sullivan as a 2017 Manufacturing Leadership Awards winner for outstanding achievement in Engineering & Production Technology Leadership. Our Lexington-based facility is one of the largest, most versatile gene therapy manufacturing facilities in the world. Over the past four years, we have invested more than $25 million in designing, constructing and equipping our 55,000 square foot facility with state-of-the-art laboratories and commercial-scale production capabilities. The facility offers 500-liter capacity with the ability to expand to up to 2,000 liters when needed.

·                   On April 4, 2017, we published data demonstrating widespread transduction in the central nervous system following direct injection of our AAV5 vector in a large animal model. The method of injection used was found to result in a controlled and accurate administration with no adverse events observed in non-human primates. Varying doses of AAV5 achieved predictable transduction of connected areas of the brain. The data demonstrate that AAV5 is an effective vector for the central nervous system and has potential for the treatment of a wide range of neurological pathologies. These data will help guide the clinical development of our gene therapy product candidate AMT-130, which consists of an AAV5 vector carrying an artificial micro-RNA that silences the huntingtin gene for the treatment of Huntington’s disease.

·                   On April 20, 2017, we announced that it will not pursue the renewal of the Glybera® (alipogene tiparvovec) marketing authorization in Europe when it is scheduled to expire on October 25, 2017.

 

23



Table of Contents

 

We will continue to supply product to Chiesi to treat any patients that are approved for treatment prior to October 25, 2017, and will be responsible for terminating the Phase IV post-approval study. According to the termination agreement executed in April 2017, we will be required to make installment payments to Chiesi totaling $2.2 million through January 2019.  Additionally, we may owe up to an additional $1.8 million depending on the number of patients treated with Glybera prior to its withdrawal, if any. Any such payments related to additional patients treated will be payable no earlier than April 1, 2019, in accordance with a schedule to be agreed upon by the parties. As a result of the withdrawal of Glybera, we expect to reduce future expenses related to the product by approximately $10 million through 2021.

 

Financial Operations Overview

 

Key components of our results of operations include the following:

 

 

 

Three months ended March 31,

 

 

 

2017

 

2016

 

 

 

in thousands

 

Total revenues

 

$

3,321

 

$

4,295

 

Research and development expenses

 

(16,994

)

(16,706

)

Selling, general and administrative expenses

 

(6,358

)

(7,298

)

Net loss

 

(20,272

)

(22,299

)

 

As of March 31, 2017, we had cash and cash equivalents of $120.3 million and $132.5 million as of December 31, 2016. We had a net loss of $20.3 million in the three months ended March 31, 2017, and $22.3 million for the same period in 2016. As of March 31, 2017, and December 31, 2016, we had accumulated deficits of $416.3 million and $396.1 million, respectively. We anticipate that our expenses will increase in the future as we:

 

·                   Advance our hemophilia B program, in collaboration with Chiesi, towards late-stage clinical development;

·                   Continue non-clinical and preclinical studies that are expected to support an IND filing for the S100A1 gene therapy program for the treatment of congestive heart failure, which is currently being developed with BMS;

·                   Continue IND-enabling studies for our proprietary Huntington’s disease gene therapy program and initiate a proof-of-concept clinical study;

·                   Advance multiple research programs related to gene therapy candidates targeting liver-directed and CNS disorders;

·                   Continue to enhance and optimize our technology platform, including our manufacturing capabilities, next-generation viral vectors and promoters, and other enabling technologies;

·                   Implement our strategic restructuring, including costs associated with consolidation of our manufacturing capabilities into our Lexington site and the termination or retention of certain employees;

·                   seek marketing approval for any product candidates that successfully complete clinical trials;

·                   acquire or in-license rights to new therapeutic targets or product candidates;

·                   enter into collaboration agreements with third parties to research, develop and/or manufacture potential product candidates;

·                   maintain, expand and protect our intellectual property portfolio, including in-licensing additional intellectual property rights from third parties;

·                   build-out our clinical, medical and regulatory capabilities in the U.S; and

·                   appoint additional executives, including a Chief Financial Officer and Chief Operating Officer.

 

See “Results of Operations” below for a discussion of the detailed components and analysis of the amounts above.

 

24



Table of Contents

 

Restructuring

 

Following the completion of our strategic review in November 2016, we announced a strategic restructuring plan aimed at refocusing our pipeline, consolidating our manufacturing operations and enhancing overall execution to drive shareholder value. As part of our plan, we intend to eliminate between 50 and 55 positions. In 2016, we accrued $1.1 million related to termination benefits offered to executive employees, expected to be paid in 2017. We also entered into termination agreements with non-executive employees in January 2017, for which the related termination benefits of $0.5 million in the aggregate will be recognized over the relevant remaining service period during 2017. These changes are expected to reduce annual operating expenses by $5.0 to $6.0 million from 2018 onwards.

 

Following our April 2017 decision not to seek extension of our European marketing authorization for Glybera after its expiration in October 2017, we will eliminate an additional 7 positions required for Glybera’s manufacturing at our Amsterdam site. We expect to incur additional termination costs of $0.3 million in 2017.

 

Critical Accounting Policies and Estimates

 

In preparing our consolidated financial statements in accordance with U.S. GAAP and pursuant to the rules and regulations promulgated by the Securities and Exchange Commission (“SEC”) we make assumptions, judgments and estimates that can have a significant impact on our net income/loss and affect the reported amounts of certain assets, liabilities, revenue and expenses, and related disclosures. On an ongoing basis, we evaluate our estimates and judgments, including those related to the BMS collaboration agreement, share-based payments, contingent consideration, valuation of derivative financial instruments, and research and development expenses. We base our estimates on historical experience, known trends and events and various other factors that are believed to be reasonable under the circumstances, the results of which form the basis for making judgments about the carrying values of assets and liabilities that are not readily apparent from other sources. Actual results may differ from these estimates under different assumptions or conditions. In making estimates and judgments, management employs critical accounting policies. During the three months ended March 31, 2017, there were no material changes to our critical accounting policies as reported in our Annual Report on Form 10-K for the year ended December 31, 2016, which was filed with the SEC on March 15, 2017.

 

Results of Operations for the Three Months Ended March 31, 2017

 

The following table presents a comparison of the three months ended March 31, 2017, and 2016.

 

 

 

Three months ended March 31,

 

 

 

2017

 

2016

 

2017 vs 2016

 

 

 

in thousands

 

Total revenues

 

$

3,321

 

$

4,295

 

$

(974

)

Operating expenses:

 

 

 

 

 

 

 

Research and development expenses

 

(16,994

)

(16,706

)

(288

)

Selling, general and administrative expenses

 

(6,358

)

(7,298

)

940

 

Total operating expenses

 

(23,352

)

(24,004

)

652

 

Other income

 

316

 

445

 

(129

)

Loss from operations

 

(19,715

)

(19,264

)

(451

)

Other non-operating items, net

 

(557

)

(2,478

)

1,921

 

Loss before income tax benefit / (expense)

 

(20,272

)

(21,742

)

1,470

 

Income tax benefit / (expense)

 

 

(557

)

557

 

Net loss

 

$

(20,272

)

$

(22,299

)

$

2,027

 

 

25



Table of Contents

 

Revenues

 

We recognize total collaboration revenues associated with development activities that are reimbursable by Chiesi and BMS under our respective collaboration agreements.

 

We recognize license revenues associated with the amortization of the non-refundable upfront payment, target designation fees and research and development milestone payments we received or might receive from Chiesi and BMS. The timing of these cash payments may differ from the recognition of revenue, as revenue is deferred and recognized over the duration of the performance period. We treat other revenue, such as sales milestone payments or service fees, as earned when receivable.

 

Our revenue for the three months ended March 31, 2017, and 2016 was as follows:

 

 

 

Three months ended March 31,

 

 

 

2017

 

2016

 

2017 vs 2016

 

 

 

in thousands

 

License revenue

 

$

1,183

 

$

1,219

 

$

(36

)

Collaboration revenue Chiesi

 

1,580

 

1,426

 

154

 

Collaboration revenue BMS

 

558

 

1,650

 

(1,092

)

Total revenues

 

$

3,321

 

$

4,295

 

$

(974

)

 

We expect to continue to recognize $1.0 million in license revenue each quarter from upfront payments and target designation fees received from BMS in the second and third quarters of 2015. We also continue to recognize $0.2 million license revenue each quarter from upfront fees received from Chiesi in 2013.

 

Collaboration revenue generated from our research activities associated with the S100A1 heart failure program on behalf of BMS during the three months ended March 31, 2017 were $0.6 million, compared to $1.7 million for the same period in 2016. The reduction in the current year period was driven by the timing of various preclinical activities as well as activities related to the production of preclinical material during the prior year period.

 

Collaboration revenue generated from our co-development of AMT-060 with Chiesi for the three months ended March 31, 2017, were $1.6 million, compared to $1.4 million for the same period in 2016.

 

Research and development expenses

 

We expense research and development costs (“R&D”) as incurred. Our R&D expenses generally consist of cost incurred for the development of our target candidates, which include:

 

·              Employee-related expenses, including salaries, benefits, travel and share-based compensation expense;

·              Costs incurred for laboratory research, preclinical and nonclinical studies, clinical trials, statistical analysis and report writing, and regulatory compliance costs incurred with clinical research organizations and other third party vendors;

·              Costs incurred to conduct consistency and comparability studies;

·              Costs incurred for the start-up and validation of our Lexington facility;

·              Costs incurred for the development and improvement of our manufacturing processes and methods;

·              Costs associated with our research activities for our next-generation vector and promoter platform;

·              Costs incurred, including share-based compensation expense, under our collaboration and license agreement with 4D Molecular Therapeutics;

·              Changes in the fair value of the contingent consideration related to our acquisition of InoCard;

·              Facilities, depreciation and other expenses, which include direct and allocated expenses for rent and maintenance of facilities, insurance and other supplies; and

·              Amortization and impairments of intangible assets.

 

26



Table of Contents

 

Our research and development expenses primarily consist of costs incurred for the research and development of our product candidates, which include:

 

·              AMT-060 ( Hemophilia B ). We initiated a Phase I/II clinical trial of AMT-060 for the treatment of hemophilia B in the first quarter of 2015 in collaboration with Chiesi. Under our co-development agreement, we and Chiesi will each bear half of the agreed development costs of this program;

·              S100A1 ( congestive heart failure ). In the third quarter of 2014, we started to incur costs related to the preclinical development of product candidates targeting the S100A1 gene. Since May 2015, all costs related to the program are reimbursed by BMS under the collaboration agreement;

·              AMT-130 ( Huntington’s disease ). We have incurred costs related to preclinical research for AMT-130;

·              AMT-110 ( Sanfilippo B ). We have incurred costs related to the development and manufacture of clinical supplies of AMT-110 for the Phase I/II clinical trial. We suspended this program in late 2016;

·              Preclinical research programs . We incur costs related to the research of multiple preclinical gene therapy product candidates with the potential to treat certain rare and other serious medical conditions; and

·              Technology platform development and other related research . We incur significant research and development costs related to vector design, manufacturing and other aspects of our modular gene therapy technology platform that are applicable across all of our programs.

 

Our research and development expenses may vary substantially from period to period based on the timing of our research and development activities, including manufacturing campaigns, regulatory submissions and enrollment of patients in clinical trials. We expect that our research and development expenses will increase significantly as we progress our preclinical and clinical programs, advance the research and development of our other product candidates and transfer manufacturing to our facility in Lexington, Massachusetts. The successful development of our product candidates is highly uncertain. At this time, we cannot reasonably estimate the nature, timing or estimated costs of, or any cash inflows resulting from, the development of any of our product candidates. This is due to numerous risks and uncertainties associated with developing gene therapies, including the uncertainty of:

 

·                       the scope, rate of progress and expense of our research and development activities;

·                       our ability to successfully manufacture and scale-up production;

·                       clinical trial protocols, speed of enrollment and resulting data;

·                       the effectiveness and safety of our product candidates;

·                       the timing of regulatory approvals; and

·                       our ability to agree to ongoing development budgets with collaborators who share the costs of our development program.

 

A change in the outcome of any of these variables with respect to our product candidates that we may develop could mean a significant change in the expenses and timing associated with the development of such product candidate.

 

Research and development expenses for the three months ended March 31, 2017, were $17.0 million, compared to $16.7 million for the same period in 2016.

 

·                       We incurred $9.0 million in personnel, share-based compensation expenses and consulting cost in the three months ended March 31, 2017, compared to $8.8 million in the three months ended March 31, 2016. The increase in 2017 is driven by one-off expenses we incur related to terminations and retention benefits as well as the addition of personnel throughout 2016;

·                       We incurred $3.9 million in external services and cost related to the development of our product candidates in the three months ended March 31, 2017, compared to $3.9 million for the same period in 2016, as we advanced our efforts to drive hemophilia B towards late-stage clinical development;

·                        We incurred $3.8 million operating expenses and depreciation expenses related to our rented facilities in the three months ended March 31, 2017, compared to $3.0 million for the same period in 2016. The increase in 2017 is driven primarily by the additional costs associated with our new Amsterdam facility, the lease for which commenced in March 2016;

 

27



Table of Contents

 

·                       We incurred no share-based compensation expenses related to our collaboration with 4D Molecular Therapeutics in the three months ended March 31, 2017, compared to $0.7 million for the same period in 2016.

 

Selling, general and administrative expenses

 

Our general and administrative expenses consist principally of employee, office, consultancy, legal and other professional and administrative expenses. We incur expenses associated with operating as a public company, including expenses for personnel, legal, accounting and audit fees, board of directors costs, directors’ and officers’ liability insurance premiums, NASDAQ listing fees, expenses related to investor relations and fees related to business development and maintaining our patent and license portfolio. Following the initiation of the commercialization of Glybera in September 2015, we incurred selling and marketing costs related to maintaining a patient registry and conducting a post-approval, Phase IV study for Glybera. These costs were recorded as research and development expenses prior to the commercialization.

 

Selling, general and administrative expenses for the three months ended March 31, 2017, were $6.4 million, compared to $7.3 million for the same period in 2016.

 

·                       Our expenses related to employees, contractors and consultants in the three months ended March 31, 2017, were $2.2 million compared to $2.4 million for the same period in 2016. The decrease was primarily related to $0.6 million in one-time costs related to the CEO-transition during the prior year period;

·                       We incurred $0.9 million of share-based compensation expenses in the three months ended March 31, 2017, compared to $1.1 million for the same period in 2016. The decrease was related to the reduction in fair value of the equity instruments offered to employees;

·                       We incurred $1.8 million of professional fees in the three months ended March 31, 2017, compared to $1.6 million for the same period in 2016. We regularly incur accounting, audit and legal fees associated with operating as a public company; and

·                       We incurred $0.2 million of costs associated with the Glybera global registry and Phase IV study as selling cost in the three months ended March 31, 2017, compared to $0.9 million for the same period in 2016. The decrease was primarily related to one-time costs to implemented post-approval requirement incurred in the first quarter 2016.

 

Other non-operating items, net

 

Our other non-operating items, net, for the three months ended March 31, 2017, and 2016 were as follows:

 

 

 

Three months ended March 31,

 

 

 

2017

 

2016

 

2017 vs 2016

 

 

 

in thousands

 

Interest income

 

$

11

 

$

22

 

$

(11

)

Interest expense Hercules long-term debt

 

(504

)

(629

)

125

 

Foreign currency gains / (losses)

 

(93

)

(2,188

)

2,095

 

Other non-operating income / (expense)

 

29

 

317

 

(288

)

Total other non-operating income / (expense), net

 

$

(557

)

$

(2,478

)

$

1,921

 

 

Our interest income consists of interest income earned on our cash and cash equivalents.

 

We hold monetary items and enter into transactions in foreign currencies, predominantly in euros and U.S. dollars. We recognize foreign exchange results related to changes in these foreign currencies. We recognized a net loss of $0.1 million in the three months ended March 31, 2017, compared to a net loss of $2.2 million for the same period in 2016 related to our borrowings from Hercules and our cash and cash equivalents.

 

We issued warrants to Hercules in 2013 and to BMS in 2015. We recognize changes in the fair value of these warrants within other non-operating income / (expense). In the three months ended March 31, 2017, we recognized a gain of $0.0 million compared to a gain of $0.3 million for the same period in 2016.

 

28



Table of Contents

 

Financial Position, Liquidity and Capital Resources

 

As of March 31, 2017, we had cash and cash equivalents of $120.3 million. We currently expect that our cash and cash equivalents will be sufficient to fund operations into 2019. The table below summarizes our consolidated cash flow data for the three months ended March 31, 2017, and 2016.

 

 

 

Three months ended March 31,

 

 

 

2017

 

2016

 

 

 

in thousands

 

Cash and cash equivalents at the beginning of the period

 

$

132,496

 

$

221,626

 

Net cash (used in) / provided by operating activities

 

(11,670

)

(15,726

)

Net cash used in investing activities

 

(1,969

)

(2,475

)

Net cash generated from financing activities

 

441

 

772

 

Foreign exchange impact

 

973

 

5,483

 

Cash and cash equivalents at the end of the period

 

$

120,271

 

$

209,680

 

 

We have incurred losses and cumulative negative cash flows from operations since our business was founded by our predecessor entity AMT Therapeutics (“AMT”) Holding N.V. in 1998. We had a net loss of $20.3 million in the three months ended March 31, 2017, compared to a loss of $22.3 million in the same period 2016. As of March 31, 2017, we had an accumulated deficit of $416.3 million.

 

Sources of liquidity

 

From our first institutional venture capital financing in 2006 through March 31, 2017, we funded our operations primarily through private and public placements of equity securities and convertible and other debt securities and to a much lesser extend upfront, target designation or similar payments from our collaboration partners as well as collaboration revenues.

 

We expect to continue to incur losses and to generate negative cash flows. We have no firm sources of additional financing other than our collaboration agreements with Chiesi and BMS and additional commitments that may be available under our Hercules loan facility. Until such time, if ever, as we can generate substantial cash flows from successfully commercializing our product candidates, we expect to finance our cash needs through a combination of equity offerings, debt financings, collaborations, strategic alliances and marketing, distribution and licensing arrangements.

 

We are subject to covenants under our Loan Agreement with Hercules, and may become subject to covenants under any future indebtedness that could limit our ability to take specific actions, such as incurring additional debt, making capital expenditures or declaring dividends, which could adversely impact our ability to conduct our business. In addition, our pledge of assets as collateral to secure our obligations under the Hercules loan agreement may limit our ability to obtain debt financing. To the extent, we need to finance our cash needs through equity offerings or debt financings, such financing may be subject to unfavorable terms including without limitation, the negotiation and execution of definitive documentation, as well as credit and debt market conditions, and we may not be able to obtain such financing on terms acceptable to us or at all. If financing is not available when needed, including through debt or equity financings, or is available only on unfavorable terms, we may be unable to meet our cash needs. If we raise additional funds through collaborations, strategic alliances or marketing, distribution or licensing arrangements with third parties, we may have to relinquish valuable rights to our technologies, future revenue streams or product candidates or grant licenses on terms that may not be favorable to us. If we are unable to raise additional funds through equity or debt financings when needed, we may be required to delay, limit, reduce or terminate our product development or future commercialization efforts or grant rights to develop and market product candidates that we would otherwise prefer to develop and market ourselves, which could have a material adverse effect on our business, financial conditions, results of operations and cash flows.

 

29



Table of Contents

 

Net Cash (used in)/generated by operating activities

 

Net cash used in operating activities was $11.7 million for the three months ended March 31, 2017, a reduction of $4.0 million compared to the $15.7 million of cash used in the same period in 2016.

 

The reduction in 2017 is due to a $3.6 million reduction of our net working capital compared to a relatively flat net working capital position during the same period in 2016. In addition, in 2017, we collected $1.1 million in lease incentive payments related to our new Amsterdam facility.

 

Net cash used in investing activities

 

In the three months ended March 31, 2017, we used $2.0 million in our investing activities compared to $2.5 million for the same period in 2016.

 

 

 

Three months ended March 31,

 

 

 

2017

 

2016

 

 

 

in thousands

 

Build out of Lexington site

 

$

(236

)

$

(691

)

Build out of Amsterdam sites

 

(1,733

)

(1,176

)

Restricted cash

 

 

(608

)

Total investments

 

$

(1,969

)

$

(2,475

)

 

In the three months ended March 31, 2017, we invested $1.7 million in our new facility in Amsterdam and $1.2 million in the same period 2016.

 

Net cash generated from financing activities

 

During the three months ended March 31, 2017, we received $0.4 million from the exercise of options to purchase common shares in relation to our share incentive plans, compared to $0.8 million in the same period 2016.

 

Funding requirements

 

We believe our cash and cash equivalents as of March 31, 2017, will enable us to fund our operating expenses including our debt repayment obligations as they become due and capital expenditure requirements, for at least the next twelve months. Our future capital requirements will depend on many factors, including but not limited to:

 

·                   the potential to receive future consideration pursuant to our collaboration with BMS, which is largely contingent on achieving certain research, development, regulatory and sales milestones;

·                   our collaboration agreements remaining in effect and our ability to enter into such new arrangements in the future;

·                   the scope, timing, and results and costs of our current and planned clinical trials, including those for hemophilia B and AMT-130 in Huntington’s Disease;

·                   the scope, timing, results and costs of preclinical development and laboratory testing of our additional product candidates, including those for the treatment of Huntington’s disease and other gene therapy product candidates;

·                   the timing and costs incurred in relation to the withdrawal of our marketing authorization for Glybera;

·                   the need for any additional tests, studies, or trials beyond those originally anticipated to confirm the safety or efficacy of our product candidates and technologies;

·                   the number of other product candidates that we pursue and their respective development requirements;

·                   the cost, timing and outcome of regulatory review of our product candidates;

·                   the cost and timing of future commercialization activities, including product manufacturing, marketing, sales and distribution, for any of our product candidates for which we receive marketing approval in the future;

·                   the amount and timing of revenue, if any, we receive from commercial sales of any product candidates for which we, or our collaboration partners, receive marketing approval in the future;

 

30



Table of Contents

 

·                   the costs and timing of preparing, filing, expanding, acquiring, licensing, maintaining, enforcing and prosecuting patents and patent applications, as well as defending any intellectual property-related claims;

·                   the repayments of the principal amount of our venture debt loan with Hercules, which will contractually start in December 2017 and will run through May 2020;

·                   the extent to which we acquire or in-license other businesses, products, product candidates or technologies;

·                   the costs associated with maintaining quality compliance and optimizing our manufacturing processes, including the operating costs associated with our Lexington, Massachusetts manufacturing facility;

·                   the costs associated with hiring additional senior management and other personnel, particularly in our clinical development, medical affairs and regulatory affairs groups;

·                   the timing, costs, savings and operational implications of the corporate restructuring we are implementing following the recent completion of our strategic review; and

·                   the costs associated with augmenting our corporate infrastructure, including the improvement of our information systems and addition of finance, human resource, legal and compliance personnel.

 

Contractual obligations and commitments

 

The table below sets forth our contractual obligations and commercial commitments as of March 31, 2017, that are expected to have an impact on liquidity and cash flows in future periods.

 

 

 

Undefined

 

Less than 1
year

 

Between 1 and
2 years

 

Between 2 and
5 years

 

Over 5 years

 

Total

 

 

 

in thousands

 

Debt obligations (including $4.4 million interest payments)

 

$

 

$

4,009

 

$

9,098

 

$

11,292

 

$

 

$

24,399

 

Operating lease obligations

 

 

2,773

 

4,016

 

11,226

 

21,668

 

39,683

 

Contingent consideration (nominal amount)

 

15,469

 

 

 

 

 

15,469

 

Total

 

$

15,469

 

$

6,782

 

$

13,114

 

$

22,518

 

$

21,668

 

$

79,551

 

 

Due to uncertainty of the timing of achieving milestones, the contingent consideration of $15.5 million related to our acquisition of InoCard (later renamed uniQure GmbH), is considered to have an undefined contractual maturity. As of March 31, 2017, we expect the milestone obligations will become payable between 2018 and 2021. When due, the obligations can be settled either in cash or in a variable number of our shares. As of March 31, 2017, we recorded this obligation at its fair value of $2.0 million.

 

We also have obligations to make future payments to third parties that become due and payable on the achievement of certain development, regulatory and commercial milestones (such as the start of a clinical trial, filing of a Biologics License Application, approval by the FDA or product launch). We have not included these commitments on our balance sheet or in the table above because the achievement and timing of these milestones is not fixed and determinable.

 

We enter into contracts in the normal course of business with CROs for preclinical research studies and clinical trials, research supplies and other services and products for operating purposes. These contracts generally provide for termination on notice, and therefore are cancelable contracts and not included in the table of contractual obligations and commitments.

 

The Company’s predecessor entity received a technical development loan from the Dutch government in relation to the development of Glybera. The Company needs to repay the grant through a percentage of revenue derived from product sales of Glybera up to December 31, 2019. Any grant balance remaining at this date will be forgiven. We have decided not to renew our marketing authorization for Glybera in the European Union, which expires on October 25, 2017. We do not expect to derive any revenue from Glybera after that time.

 

Off-Balance Sheet Arrangements

 

As of March 31, 2017, we did not have any off-balance sheet arrangements as defined in Item 303(a) (40) of Regulation S-K.

 

31



Table of Contents

 

Item 3. Quantitative and Qualitative Disclosures about Market Risk

 

We are exposed to a variety of financial risks in the normal course of our business, including market risk (including currency, price and interest rate risk), credit risk and liquidity risk. Our overall risk management program focuses on preservation of capital and the unpredictability of financial markets and has sought to minimize potential adverse effects on our financial performance and position.

 

Our market risks and exposures to such market risks during the three months ended March 31, 2017, has not materially changed from our market risks and our exposure to market risk discussed in Part II, Item 7A of our Annual Report on Form 10-K for the year ended December 31, 2016, which was filed with the SEC on March 15, 2017.

 

Item 4. Controls and Procedures

 

Evaluation of Disclosure Controls and Procedures

 

Our management, with the participation of our chief executive and interim finance officer (“CEO”), evaluated the effectiveness of our disclosure controls and procedures (as defined in Rules 13a-15(e) and 15d-15(e) under the Securities Exchange Act of 1934, as amended (the “Exchange Act”)) as of March 31, 2017. Based on such evaluation, our CEO has concluded that as of March 31, 2017, our disclosure controls and procedures were effective to ensure that information required to be disclosed by it in reports the Company files or submits under the Exchange Act is recorded, processed, summarized and reported within the time periods specified in the rules and forms of the SEC, and that such material information is accumulated and communicated to the Company’s management, including its Principal Executive Officer and Principal Financial Officer, to allow timely decisions regarding required disclosure. Because of the inherent limitations in all control systems, any controls and procedures, no matter how well designed and operated, can provide only reasonable assurance of achieving the desired control objectives, and management necessarily is required to apply its judgment in evaluating the cost-benefit relationship of possible controls and procedures. Furthermore, the Company’s controls and procedures can be circumvented by the individual acts of some persons, by collusion of two or more people or by management override of such control, and misstatements due to error or fraud may occur and not be detected on a timely basis.

 

Changes in Internal Control over Financial Reporting

 

During the three months ended March 31, 2017, there were no changes in our internal control over financial reporting (as defined in Rule 13a-15(f) under the Exchange Act) that materially affected, or are reasonably likely to affect, internal control over financial reporting.

 

32



Table of Contents

 

Part II — OTHER INFORMATION

 

Item 1. Legal Proceedings

 

None.

 

Item 1A. Risk Factors

 

An investment in our common shares involves a high degree of risk. You should carefully consider the following information about these risks, together with the other information appearing elsewhere in this Quarterly Report on Form 10-Q, including our financial statements and related notes thereto, and the risk factors discussed in Part I, Item 1A “Risk Factors” in our Annual Report on Form 10-K for the year ended December 13, 2016, field with the SEC on March 15, 2017, before deciding to invest in our common shares. We operate in a dynamic and rapidly changing industry that involves numerous risks and uncertainties. The risks and uncertainties described below are not the only ones we face. Other risks and uncertainties, including those that we do not currently consider material, may impair our business. If any of the risks discussed below actually occur, our business, financial condition, operating results or cash flows could be materially adversely affected. This could cause the value of our securities to decline, and you may lose all or part of your investment.

 

Those risk factors below denoted with a “*” are newly added or have been materially updated from our Annual Report on 10-K filed with the SEC on March 15, 2017.

 

Risks Related to the Development of Our Product Candidates

 

We may encounter substantial delays in and impediments to the progress of our clinical trials or fail to demonstrate the safety and efficacy of our product candidates.

 

Clinical and non-clinical development is expensive, time-consuming and uncertain as to outcome. Our product candidates are in early clinical or preclinical development, and there is a significant risk of failure or delay in each of these programs. We cannot guarantee that any preclinical tests or clinical trials will be completed as planned or completed on schedule, if at all. A failure of one or more preclinical tests or clinical trials can occur at any stage of testing. Events that may prevent successful or timely completion of clinical development include, but are not limited to:

 

·                       delays in reaching a consensus with regulatory agencies on study design;

·                       delays in reaching agreement on acceptable terms with prospective clinical research organizations (“CROs”) and clinical trial sites;

·                       delays in receiving regulatory authority to conduct the clinical trials or a regulatory authority decision that the clinical trial should not proceed;

·                       delays in obtaining required Institutional Review Board (“IRB”) approval at each clinical trial site;

·                       imposition of a clinical hold by regulatory agencies after an inspection of our clinical trial operations or trial sites;

·                       failure by CROs, other third parties or us to adhere to clinical trial requirements or otherwise properly manage the clinical trial process, including meeting applicable timelines, properly documenting case files, including the retention of proper case files, and properly monitoring and auditing clinical sites;

·                       failure of sites or clinical investigators to perform in accordance with good clinical practices (“GCP”) or applicable regulatory guidelines in other countries;

·                       difficulty or delays in patient recruiting into clinical trials;

·                       delays or deviations in the testing, validation, manufacturing and delivery of our product candidates to the clinical sites;

·                       delays in having patients complete participation in a study or return for post-treatment follow-up;

·                       clinical trial sites or patients dropping out of a study;

·                       occurrence of serious adverse events associated with a product candidate that are viewed to outweigh its potential benefits; or

·                       changes in regulatory requirements and guidance that require amending or submitting new clinical protocols, undertaking additional new tests or analyses or submitting new types or amounts of clinical data.

 

33



Table of Contents

 

Before obtaining marketing approval from regulatory authorities for the sale of our product candidates, we must conduct extensive clinical trials to demonstrate the safety and efficacy of the product candidates in humans. Such trials and regulatory review and approval take many years. It is impossible to predict when or if any of our clinical trials will demonstrate that product candidates are effective or safe in humans. If the results of our clinical trials are inconclusive, or fail to meet the level of statistical significance required for approval or if there are safety concerns or adverse events associated with our product candidates, we may:

 

·                       be delayed in or altogether prevented from obtaining marketing approval for our product candidates;

·                       obtain approval for indications or patient populations that are not as broad as intended or desired;

·                       obtain approval with labeling that includes significant use or distribution restrictions or safety warnings;

·                       be subject to changes with the way the product is administered;

·                       be required to perform additional clinical trials to support approval or be subject to additional post-marketing testing requirements;

·                       have regulatory authorities withdraw their approval of the product or impose restrictions on its distribution in the form of a modified risk evaluation and mitigation strategy;

·                       be subject to the addition of labeling statements, such as warnings or contraindications;

·                       be sued; or

·                       experience damage to our reputation.

 

Because of the nature of the gene therapies we are developing, regulators may also require us to demonstrate long-term gene expression or clinical efficacy, which may require additional or longer clinical trials and which may not be able to be demonstrated to the regulatory authorities’ standards.

 

Our ability to recruit patients for our trials is often reliant on third parties, such as the pharmacies at our clinical trial sites. These third parties may not have the adequate infrastructure established to handle gene therapy products or to support certain gene therapy product formulations, or may not agree to recruit patients on our behalf.

 

In addition, we or our collaborators may not be able to locate and enroll a sufficient number of eligible patients to participate in these trials as required by the FDA, the EMA or similar regulatory authorities outside the United States and the European Union. This may result in our failure to initiate or continue clinical trials for our product candidates, or may cause us to abandon one or more clinical trials altogether. In particular, because several of our programs are focused on the treatment of patients with rare, orphan or ultra-orphan diseases, our ability to enroll eligible patients in these trials may be limited or slower than we anticipate in light of the small patient populations involved and the specific age range required for treatment eligibility in some indications. In addition, our potential competitors, including major pharmaceutical, specialty pharmaceutical and biotechnology companies, academic institutions and governmental agencies and public and private research institutions, may seek to develop competing therapies, which would further limit the small patient pool available for our studies.

 

Any inability to successfully initiate or complete preclinical and clinical development could result in additional costs to us or impair our ability to receive marketing approval, to generate revenues from product sales or obtain regulatory and commercialization milestones and royalties. In addition, if we make manufacturing or formulation changes to our product candidates, including changes in the vector or manufacturing process used, we may need to conduct additional studies to bridge our modified product candidates to earlier versions. Clinical trial delays could also shorten any periods during which we may have the exclusive right to commercialize our product candidates or allow our competitors to bring products to market before we do, which could impair our ability to successfully commercialize our product candidates and may harm our business and results of operations.

 

Our progress in early-stage clinical trials may not be indicative of long-term efficacy in late-stage clinical trials, and our progress in trials for one product candidate may not be indicative of progress in trials for other product candidates.

 

The product candidates in our pipeline are at early-stages of development. Study designs and results from previous studies are not necessarily predictive of our future clinical study designs or results, and initial results may not be confirmed upon full analysis of the complete study data. Our product candidates may fail to show the required level of safety and efficacy in later stages of clinical development despite having successfully advanced through initial clinical studies.

 

34



Table of Contents

 

A number of companies in the pharmaceutical and biotechnology industries have suffered significant setbacks in late-stage clinical trials even after achieving promising results in early-stage clinical trials. If a larger population of patients does not experience positive results during clinical trials, if these results are not reproducible or if our products show diminishing activity over time, our products may not receive approval from the FDA or EMA. Data obtained from preclinical and clinical activities are subject to varying interpretations, which may delay, limit or prevent regulatory approval. In addition, we may encounter regulatory delays or rejections as a result of many factors, including changes in regulatory policy during the period of product development. Failure to confirm favorable results from earlier trials by demonstrating the safety and effectiveness of our products in late-stage clinical trials with larger patient populations could have a material adverse effect on our business that would cause our share price to decline.

 

Fast track product, breakthrough therapy, priority review, or Regenerative Advanced Therapy (“RAT”) designation by the FDA, or access to the PRIME scheme by the EMA, for our product candidates may not lead to faster development or regulatory review or approval process, and it does not increase the likelihood that our product candidates will receive marketing approval.

 

We may seek fast track, a breakthrough therapy designation, RAT designation, and priority review designation and PRIME scheme access for our product candidates if supported by the results of clinical trials. A fast track product designation is designed to facilitate the clinical development and expedite the review of drugs intended to treat a serious or life-threatening condition which demonstrate the potential to address an unmet medical need. A breakthrough therapy is defined as a drug that is intended, alone or in combination with one or more other drugs, to treat a serious or life-threatening disease or condition, and preliminary clinical evidence indicates that the drug may demonstrate substantial improvement over existing therapies on one or more clinically significant endpoints, such as substantial treatment effects observed early in clinical development. A RAT designation is designed to accelerate approval for regenerative advanced therapies. Priority review designation is intended to speed the FDA marketing application review timeframe for drugs that treat a serious condition and, if approved, would provide a significant improvement in safety or effectiveness. PRIME is a scheme provided by the EMA to enhance support for the development of medicines that target an unmet medical need.

 

For drugs and biologics that have been designated as fast track products or breakthrough therapies, or granted access to the PRIME schema, interaction and communication between the regulatory agency and the sponsor of the trial can help to identify the most efficient path for clinical development. Sponsors of drugs with fast track products or breakthrough therapies may also be able to submit marketing applications on a rolling basis, meaning that the FDA may review portions of a marketing application before the sponsor submits the complete application to the FDA, as long as the sponsor pays the user fee upon submission of the first portion of the marketing application. For products that receive a priority review designation, the FDA’s marketing application review goal is shortened to six months, as opposed to ten months under standard review. This review goal is based on the date the FDA accepts the marketing application for review, which typically adds approximately two months to the timeline for review and decision from the date of submission. RAT designations will accelerate approval but the exact mechanisms have not yet been announced by FDA.

 

Designation as a fast track product, breakthrough therapy, RAT, PRIME, or priority review product is within the discretion of the regulatory agency. Accordingly, even if we believe one of our product candidates meets the criteria for designation as a fast track product, breakthrough therapy, RAT, PRIME, or priority review product, the agency may disagree and instead determine not to make such designation. In any event, the receipt of such a designation for a product candidate may not result in a faster development process, review or approval compared to drugs considered for approval under conventional regulatory procedures and does not assure ultimate marketing approval by the agency. In addition, with regard to fast track products and breakthrough therapies, the FDA may later decide that the products no longer meet the conditions for qualification as either a fast track product, RAT, or a breakthrough therapy or, for priority review products, decide that the time period for FDA review or approval will not be shortened.

 

35



Table of Contents

 

We may not be successful in our efforts to use our gene therapy technology platform to build a pipeline of additional product candidates.

 

An element of our strategy is to use our gene therapy technology platform to expand our product pipeline and to progress these candidates through clinical development ourselves or together with our collaborators. Although we currently have a pipeline of programs at various stages of development, we may not be able to identify or develop product candidates that are safe and effective. Even if we are successful in continuing to build our pipeline, the potential product candidates that we identify may not be suitable for clinical development. Research programs to identify new product candidates require substantial technical, financial and human resources. We or our collaborators may focus our efforts and resources on potential programs or product candidates that ultimately prove to be unsuccessful. If we do not continue to successfully develop and commercialize product candidates based upon our technology, we may face difficulty in obtaining product revenues in future periods, which could result in significant harm to our financial position and adversely affect our share price.

 

Our strategy of obtaining rights to key technologies through in-licenses may not be successful.

 

We seek to expand our product pipeline in part by in-licensing the rights to key technologies, including those related to gene delivery, genes and gene cassettes. The future growth of our business will depend in significant part on our ability to in-license or otherwise acquire the rights to additional product candidates or technologies, particularly through our collaborations with academic research institutions. However, we may be unable to in-license or acquire the rights to any such product candidates or technologies from third parties on acceptable terms or at all. The in-licensing and acquisition of these technologies is a competitive area, and a number of more established companies are also pursuing strategies to license or acquire product candidates or technologies that we may consider attractive. These established companies may have a competitive advantage over us due to their size, cash resources and greater clinical development and commercialization capabilities. In addition, companies that perceive us to be competitors may be unwilling to license rights to us. Furthermore, we may be unable to identify suitable product candidates or technologies within our areas of focus. If we are unable to successfully obtain rights to suitable product candidates or technologies, our business, financial condition and prospects could suffer.

 

Negative public opinion and increased regulatory scrutiny of gene therapy and genetic research may damage public perception of our product candidates or adversely affect our ability to conduct our business or obtain marketing approvals for our product candidates. *

 

Public perception may be influenced by claims that gene therapy is unsafe, and gene therapy may not gain the acceptance of the public or the medical community. The risk of cancer remains a concern for gene therapy, and we cannot assure that it will not occur in any of our planned or future clinical studies. In addition, there is the potential risk of delayed adverse events following exposure to gene therapy products due to persistent biological activity of the genetic material or other components of products used to carry the genetic material.

 

As of December 31, 2016, a total of two serious adverse event reports in AMT-060-treated patients occurred in our Phase I/II hemophilia B trial, a transient elevation of liver transaminases and a fever, which were assessed as probably, and possibly related to AMT-060, respectively.

 

Adverse events in our clinical trials or those conducted by other parties (even if not ultimately attributable to our product candidates), and the resulting publicity, could result in increased governmental regulation, unfavorable public perception, failure of the medical community to accept and prescribe gene therapy treatments, potential regulatory delays in the testing or approval of our product candidates, stricter labeling requirements for those product candidates that are approved and a decrease in demand for any such product candidates.

 

36



Table of Contents

 

Risks Related to Our Manufacturing

 

Delays in qualifying our U.S. manufacturing facility for clinical production activities could delay our development and commercialization plans and thereby limit our revenues and growth. *

 

We have commenced consolidating all manufacturing at our facility in Lexington, Massachusetts, which is currently in the qualification process to enable production of clinical trial material. If qualification is delayed, we may not be able to manufacture sufficient quantities of our product candidates, which would limit our commercialization and development activities and our opportunities for growth. Cost overruns associated with this facility could also require us to raise additional funds from external sources, which may be unavailable on favorable terms or at all.

 

Our manufacturing facility is subject to significant government regulations and approvals. If we fail to comply with these regulations or maintain these approvals our business will be materially harmed. *

 

Our manufacturing facility in Lexington will be subject to ongoing regulation and periodic inspection by the EMA, FDA and other regulatory bodies to ensure compliance with current Good Manufacturing Practices (“cGMP”). Any failure to follow and document our adherence to such cGMP regulations or other regulatory requirements may lead to significant delays in the availability of products for commercial sale or clinical study, may result in the termination of or a hold on a clinical study, or may delay or prevent filing or approval of marketing applications for our products.

 

Failure to comply with applicable regulations could also result in the EMA, FDA or other applicable authorities taking various actions, including levying fines and other civil penalties; imposing consent decrees or injunctions; requiring us to suspend or put on hold one or more of our clinical trials; suspending or withdrawing regulatory approvals; delaying or refusing to approve pending applications or supplements to approved applications; requiring us to suspend manufacturing activities or product sales, imports or exports; requiring us to communicate with physicians and other customers about concerns related to actual or potential safety, efficacy, and other issues involving our products; mandating product recalls or seizing products; imposing operating restrictions; and seeking criminal prosecutions. Any of the foregoing could materially harm our business.

 

Gene therapies are complex and difficult to manufacture. We could experience production or technology transfer problems that result in delays in our development or commercialization schedules or otherwise adversely affect our business.

 

We manufacture our clinical and commercial supplies of our product candidates ourselves in our GMP certified facility in Amsterdam. We plan to decommission this facility in the second quarter of 2017 and have begun transferring all manufacturing to our facility in Lexington, Massachusetts, which will involve a complex process of technology transfer. The insect-cell based manufacturing process we use to produce our products and product candidates is highly complex and in the normal course is subject to variation or production difficulties. Issues with the manufacturing process, even minor deviations from the normal process, could result in insufficient yield, product deficiencies or manufacturing failures that result in lot failures, insufficient inventory, product recalls and product liability claims. We may encounter problems in completing our technology transfer or in achieving adequate or clinical-grade materials that meet EMA, FDA or other applicable standards or specifications with consistent and acceptable production yields and costs.

 

A number of factors common to the manufacturing of most biologics and drugs could also cause production interruptions, including raw materials shortages, raw material failures, growth media failures, equipment malfunctions, facility contamination, labor problems, natural disasters, disruption in utility services, terrorist activities, or acts of god beyond our control. We also may encounter problems in hiring and retaining the experienced specialized personnel needed to operate our manufacturing process, particularly as we transition manufacturing to Lexington, which could result in delays in our production or difficulties in maintaining compliance with applicable regulatory requirements.

 

Any problems in our manufacturing processes or facilities could make us a less attractive collaborator for academic research institutions and other parties, which could limit our access to additional attractive development programs, result in delays in our clinical development or marketing schedules and harm our business.

 

37



Table of Contents

 

Our use of viruses, chemicals and other hazardous materials requires us to comply with regulatory requirements and exposes us to significant potential liabilities.

 

Our development and manufacturing processes involve the use of viruses, chemicals, other (potentially) hazardous materials and produce waste products. Accordingly, we are subject to national, federal, state and local laws and regulations in the United States, the Netherlands and Germany governing the use, manufacture, distribution, storage, handling, treatment and disposal of these materials. In addition to ensuring the safe handling of these materials, applicable requirements require increased safeguards and security measures for many of these agents, including controlling access and screening of entities and personnel who have access to them, and establishing a comprehensive national database of registered entities. In the event of an accident or failure to comply with environmental, occupational health and safety and export control laws and regulations, we could be held liable for damages that result, and any such liability could exceed our assets and resources.

 

Risks Related to Regulatory Approval

 

We cannot predict when or if we will obtain marketing approval to commercialize a product candidate *

 

The development and commercialization of our product candidates, including their design, testing, manufacture, safety, efficacy, purity, recordkeeping, labeling, storage, approval, advertising, promotion, sale and distribution, are subject to comprehensive regulation by the FDA and other regulatory agencies in the United States, the EMA and other regulatory agencies of the member states of the European Union, and similar regulatory authorities in other jurisdictions. Failure to obtain marketing approval for a product candidate in a specific jurisdiction will prevent us from commercializing the product candidate in that jurisdiction.

 

The process of obtaining marketing approval for our product candidates in the European Union, the United States and other countries is expensive and may take many years, if approval is obtained at all. Changes in marketing approval policies during the development period, changes in or the enactment of additional statutes or regulations, or changes in regulatory review for each submitted product application, may cause delays in the approval or rejection of an application. Regulatory authorities have substantial discretion in the approval process and may refuse to accept any application, may decide that our data are insufficient for approval, may require additional preclinical, clinical or other studies and may not complete their review in a timely manner. Further, any marketing approval we ultimately obtain may be for only limited indications, or be subject to stringent labeling or other restrictions or post-approval commitments that render the approved product not commercially viable.

 

If we experience delays in obtaining marketing approval of any of our product candidates in the United States or other countries, the commercial prospects of our other product candidates may be harmed and our ability to generate revenues will be materially impaired.

 

The FDA will require us to conduct comparability studies evaluating the products manufactured at our Amsterdam facility with those to be manufactured at our new Lexington, Massachusetts facility. Those studies and their results could substantially delay or preclude our ability to commercialize our product candidates in the United States.

 

The FDA maintains strict requirements governing the manufacturing process for biologics. When a manufacturer seeks to modify or change that process, or begin manufacturing at a new facility, the FDA typically requires the applicant to conduct non-clinical and, depending on the magnitude of the changes, potentially clinical comparability studies that evaluate the potential differences in the product resulting from the change in the manufacturing process or facility. In connection with any application for marketing approval in the United States, we will be required to conduct comparability studies assessing product manufactured at our facility in Amsterdam with product to be manufactured at our new facility in Lexington, Massachusetts.

 

Delays in designing and completing a comparability study to the satisfaction of the FDA could delay or preclude our development and commercialization plans and, thereby, increase the risk and time to achieve regulatory approval. For example, we may attempt to show comparability of the product manufactured at our Amsterdam and Lexington facilities through the use of non-clinical data, such as potency assays and animal studies. In the event that the FDA does not accept such non-clinical comparability data, we may need to conduct additional studies involving dosing of animals or patients. These potential studies may result in a delay of the approval or launch of product in the United States.

 

38



Table of Contents

 

The risks associated with the marketing approval process are heightened by the status of our products as gene therapies.

 

We believe that all our current product candidates will be viewed as gene therapy products by the applicable regulatory authorities. Gene therapies are relatively new treatments for which regulators do not have extensive experience or standard review and approval processes. The FDA unlike the EMA, does not have an exceptional circumstances approval pathway.

 

Both the FDA and EMA have demonstrated caution in their regulation of gene therapy treatments, and ethical and legal concerns about gene therapy and genetic testing may result in additional regulations or restrictions on the development and commercialization of our product candidates that are difficult to predict. The FDA and the EMA have issued various guidance documents pertaining to gene therapy products, with which we likely must comply to gain regulatory approval of any of our product candidates in the United States or European Union, respectively. The close regulatory scrutiny of gene therapy products may result in delays and increased costs, and may ultimately lead to the failure to obtain approval for any gene therapy product.

 

Regulatory requirements affecting gene therapy have changed frequently and may continue to change, and agencies at both the U.S. federal and state level, as well as congressional committees and foreign governments, have sometimes expressed interest in further regulating biotechnology. For example, the European Commission conducted a public consultation in early 2013 on the application of EU legislation that governs advanced therapy medicinal products, including gene therapy products, which could result in changes in the data we need to submit to the EMA in order for our product candidates to gain regulatory approval or change the requirements for tracking, handling and distribution of the products which may be associated with increased costs. In addition, divergent scientific opinions among the various bodies involved in the review process may result in delays, require additional resources and ultimately result in rejection. These regulatory agencies, committees and advisory groups and the new regulations and guidelines they promulgate may lengthen the regulatory review process, require us to perform additional studies, increase our development costs, lead to changes in regulatory positions and interpretations, delay or prevent approval and commercialization of our product candidates or lead to significant post-approval limitations or restrictions. Delay or failure to obtain, or unexpected costs in obtaining, the regulatory approval necessary to bring a potential product to market could decrease our ability to generate sufficient product revenues to maintain our business.

 

Our failure to obtain or maintain orphan product exclusivity for any of our product candidates for which we seek this status could limit our commercial opportunity, and if our competitors are able to obtain orphan product exclusivity before we do, we may not be able to obtain approval for our competing products for a significant period of time.

 

Regulatory authorities in some jurisdictions, including the European Union and the United States, may designate drugs for relatively small patient populations as orphan drugs. Generally, if a product with an orphan drug designation subsequently receives the first marketing approval for the relevant indication, the product is entitled to a period of market exclusivity, which precludes the EMA or FDA from approving another marketing application for the same drug for the same indication for that time period. The EMA, however, may subsequently approve a similar drug for the same indication during the first product’s market exclusivity if the EMA concludes that the later drug is clinically superior in that it is shown to be safer, more effective or makes a major contribution to patient care.

 

Orphan drug exclusivity may be lost if the EMA or FDA determines that the request for designation was materially defective, or if the manufacturer is unable to assure sufficient quantity of the drug to meet the needs of patients with the rare disease or condition or if the incidence and prevalence of patients who are eligible to receive the drug in these markets materially increase.

 

39



Table of Contents

 

As appropriate, we intend to seek all available periods of regulatory exclusivity for our product candidates. However, there is no guarantee that we will be granted these periods of regulatory exclusivity or that we will be able to maintain these periods of exclusivity.

 

The FDA grants product sponsors certain periods of regulatory exclusivity, during which the agency may not approve, and in certain instances, may not accept, certain marketing applications for competing drugs. For example, biologic product sponsors may be eligible for twelve years of exclusivity from the date of approval, seven years of exclusivity for drugs that are designated to be orphan drugs, and/or a six-month period of exclusivity added to any existing exclusivity period or patent life for the submission of FDA requested pediatric data. While we intend to apply for all periods of market exclusivity that we may be eligible for, there is no guarantee that we will receive all such periods of market exclusivity. Additionally, under certain circumstances, the FDA may revoke the period of market exclusivity. Thus, there is no guarantee that we will be able to maintain a period of market exclusivity, even if granted. In the case of orphan designation, other benefits, such as tax credits and exemption from user fees may be available. If we are not able to obtain or maintain orphan drug designation or any period of market exclusivity to which we may be entitled, we will be materially harmed, as we will potentially be subject to greater market competition and may lose the benefits associated with programs.

 

The termination of our Commercialization Agreement with Chiesi and our efforts to withdraw the marketing authorization for Glybera in Europe may expose us to financial and other risks*

 

On April 20, 2017, we announced that we agreed to terminate our Glybera Agreement with Chiesi and will not seek a marketing authorization renewal for Glybera in Europe. Under the terms of the termination of the Commercialization Agreement, we are obligated to make certain payments to Chiesi and are responsible for bearing all costs associated with certain activities associated with the winding down of the Glybera program. We are also obligated to continue to provide Glybera to Chiesi for the treatment of patients until October 25, 2017. While we believe that the financial obligations under the termination agreement are not material to the Company, the Company may be forced to incur additional costs in connection with the termination and our related obligations. We may also have to devote our resources to the winding down of the Glybera program in the period prior to termination, which may prevent us from applying our resources to other programs.

 

Risks Related to Commercialization

 

If we or our collaborators are unable to successfully commercialize our product candidates or experience significant delays in doing so, our business will be materially harmed.

 

Our ability to generate product revenues will depend on the successful development and eventual commercialization of our product candidates. The success of our product candidates will depend on a number of factors, including:

 

·                   successful completion of preclinical studies and clinical trials;

·                   receipt and maintenance of marketing approvals from applicable regulatory authorities;

·                   our ability to timely manufacture sufficient quantities according to required quality specifications;

·                   obtaining and maintaining patent and trade secret protection and non-patent, orphan drug exclusivity for our product candidates;

·                   obtaining and maintaining regulatory approval for our manufacturing facility in Lexington, Massachusetts;

·                   launch and commercialization of our products, if and when approved, whether alone or in collaboration with others;

·                   identifying and engaging effective distributors or resellers on acceptable terms in jurisdictions where we plan to utilize third parties for the marketing and sales of our product candidates;

·                   acceptance of our products, if and when approved, by patients, the medical community and third party payers;

·                   effectively competing with existing therapies and gene therapies based on safety and efficacy profile;

·                   achieve value based pricing levels based on durability of expression, safety and efficacy;

·                   obtaining and maintaining healthcare coverage and adequate reimbursement; and

 

40



Table of Contents

 

·                   complying with any applicable post-approval requirements and maintaining a continued acceptable overall safety profile.

 

Failure to achieve or implement any of these elements could result in significant delays or an inability to successfully commercialize our product candidates, which could materially harm our business.

 

The affected populations for our gene therapies may be smaller than we or third parties currently project, which may affect the size of our addressable markets.

 

Our projections of the number of people who have the diseases we are seeking to treat, as well as the subset of people with these diseases who have the potential to benefit from treatment with our therapies, are estimates based on our knowledge and understanding of these diseases. The total addressable market opportunities for these therapies will ultimately depend upon a number of factors, including the diagnosis and treatment criteria included in the final label, if approved for sale in specified indications, acceptance by the medical community, patient consent, patient access and product pricing and reimbursement. For example, after obtaining marketing authorization for Glybera from the EMA in 2013, various national European authorities denied reimbursement under national insurance schemes.

 

Prevalence estimates are frequently based on information and assumptions that are not exact and may not be appropriate, and the methodology is forward-looking and speculative. The use of such data involves risks and uncertainties and is subject to change based on various factors. Our estimates may prove to be incorrect and new studies may change the estimated incidence or prevalence of the diseases we seek to address. The number of patients with the diseases we are targeting may turn out to be lower than expected or may not be otherwise amenable to treatment with our products, or new patients may become increasingly difficult to identify or access, any of which would adversely affect our results of operations and our business.

 

The addressable market for AAV-based gene therapies are also impacted by the prevalence of neutralizing antibodies to the capsids, which are an integral component of our gene therapy constructs. Patients that have pre-existing antibodies to a particular capsid are generally not eligible for administration of a gene therapy that includes this particular capsid. For example, our AMT-060 gene therapy candidate for hemophilia B patients incorporates an AAV5 capsid. In our Phase I/II clinical study, we screened patients for preexisting anti-AAV5 antibodies in order to determine their eligibility for the trial. While none of the 10 patients screened for the study tested positive for anti-AAV5 antibodies, we have limited clinical and preclinical data on the prevalence of anti-AAV5 antibodies, and it is possible that future clinical studies may demonstrate a higher prevalence of anti-AAV5 antibodies in hemophilia B patients. This may limit the addressable market for AMT-060 and any future revenues derived from the sale of the product.

 

Any approved gene therapy we seek to offer may fail to achieve the degree of market acceptance by physicians, patients, third party payers and others in the medical community necessary for commercial success.

 

Doctors may be reluctant to accept a gene therapy as a treatment option or, where available, choose to continue to rely on existing symptomatic treatments. The degree of market acceptance of any of our product candidates that receive marketing approval in the future will depend on a number of factors, including:

 

·                   the efficacy and potential advantages of our therapies compared with alternative treatments;

·                   our ability to convince payers of the long-term cost-effectiveness of our therapies and, consequently, the availability of third party coverage and adequate reimbursement;

·                   the limitations on use and label requirements imposed by regulators;

·                   the convenience and ease of administration of our gene therapies compared with alternative treatments;

·                   the willingness of the target patient population to try new therapies, especially a gene therapy, and of physicians to administer these therapies;

·                   the strength of marketing and distribution support;

·                   the prevalence and severity of any side effects;

·                   limited access to site of service that can perform the product preparation and administer the infusion; and

·                   any restrictions on the use of our products.

 

41



Table of Contents

 

We face substantial competition, which may result in others discovering, developing or commercializing products before or more successfully than we do.

 

The development and commercialization of new biotechnology and biopharmaceutical products, including gene therapies, is highly competitive. We may face competition with respect to our product candidates, as well as with respect to any product candidates that we may seek to develop or commercialize in the future, from large and specialty pharmaceutical companies and biotechnology companies worldwide, who currently market and sell products or are pursuing the development of products for the treatment of many of the disease indications for which we are developing our product candidates. Potential competitors also include academic institutions, government agencies and other public and private research organizations that conduct research, seek patent protection and establish collaborative arrangements for research, development, manufacturing and commercialization. In recent years, there has been a significant increase in commercial and scientific interest and financial investment in gene therapy as a therapeutic approach, which has intensified the competition in this area.

 

We are aware of numerous companies focused on developing gene therapies in various indications, including AGTC, Abeona Therapeutics, Adverum Biotechnologies, Asklepios BioPharmaceutical, Audentes Therapeutics, Avalanche Biotech, AveXis, Bayer, BioMarin, Bioveratiy, bluebird bio, Dimension Therapeutics, Errant Gene Therapeutics, Expression Therapeutics, Freeline Therapeutics, Genethon, Genzyme, GlaxoSmithKline, Homology Medicines, Lysogene, Megenics, Milo Therapeutics, Nightstarx, Pfizer, REGENXBIO, Renova Therapeutics, Retrosense Therapeutics, Sangamo BioSciences, Shire, Solid Biosciences, Spark Therapeutics, Takara, and Voyager, as well as several companies addressing other methods for modifying genes and regulating gene expression. We may also face competition with respect to the treatment of some of the diseases that we are seeking to target with our gene therapies from protein pharmaceuticals under development at pharmaceutical and biotechnology companies such as Amgen, Bayer, Biogen, BioMarin, Genzyme, Novartis, Novo Nordisk, Pfizer, Shire, and numerous other pharmaceutical and biotechnology firms.

 

Our commercial opportunity could be reduced or eliminated if our competitors develop and commercialize products that are safer, more effective, have fewer or less severe side effects, are more convenient or are less expensive than the products that we develop. Our competitors also may obtain FDA, EMA or other regulatory approval for their products more rapidly than we do, which could result in our competitors establishing a strong market position before we are able to enter the market. Because we expect that gene therapy patients may generally require only a single administration, we believe that the first gene therapy product to enter the market for a particular indication will likely enjoy a significant commercial advantage, and may also obtain market exclusivity under applicable orphan drug regimes.

 

Many of the companies with which we are competing or may compete in the future have significantly greater financial resources and expertise than we do in research and development, manufacturing, preclinical testing, conducting clinical trials, obtaining regulatory approvals and marketing approved products. Mergers and acquisitions in the pharmaceutical and biotechnology industries may result in more resources being concentrated among a smaller number of our competitors. Smaller and other early-stage companies may also prove to be significant competitors, particularly through collaborative arrangements with large and established companies. These third parties compete with us in recruiting and retaining qualified scientific and management personnel, establishing clinical trial sites and patient registration for clinical trials, as well as in acquiring technologies complementary to, or necessary for, our programs.

 

Risks Related to Our Dependence on Third Parties

 

If our collaboration with BMS is not successful or if BMS designates or develops fewer targets than permitted under our collaboration agreement, our development plans, financial position and opportunities for growth may be adversely affected.

 

In order to earn all milestone payments and royalties potentially due under our collaboration with BMS, we are dependent on BMS electing to designate and actively pursue target indications covered by the collaboration and our achievement of all development, clinical and regulatory milestones under the collaboration. If BMS designates or actively pursues fewer development targets, utilizes contract research organizations, instead of our organization, to conduct non-clinical and pre-clinical studies, or if we fail to

 

42



Table of Contents

 

achieve a significant number of the applicable milestones, the total payments we receive under this collaboration may be materially lower than are potentially payable.

 

In connection with our 2014 acquisition of the InoCard business (later renamed uniQure GmbH), we undertook certain obligations regarding the development of the acquired program pursuant to a plan to be agreed between us and the sellers. The acquisition agreement provides that, in the case of an unremedied breach by us of these development obligations, the sellers could be entitled, in defined circumstances, to repurchase the InoCard business from us. If we were to breach such development obligations and were not successful in remedying such breach, the sellers might seek to exercise this repurchase right or to claim other financial penalties. Although we are diligently pursuing the development of the acquired program through our collaboration with BMS, and believe that we have not breached and will not breach such development obligations, any claim of breach could result in distraction of management and staff attention. In the event that the sellers were successful in pursuing a claim of breach by us of such obligations, our financial position and our efforts to develop S100A1 together with our collaboration partner BMS could be materially adversely affected.

 

We rely on third parties for important aspects of our development programs. If these parties do not perform successfully or if we are unable to maintain any of our collaboration arrangements, or enter into new collaborations, our business could be adversely affected.

 

We have entered into, and expect in the future to enter into, collaborations with other companies and academic research institutions with respect to important elements of our commercial and development programs. For example, we have collaboration agreements with BMS for the development and commercialization of gene therapies for cardiovascular and potentially other diseases and with Chiesi, for the co-development and commercialization of our hemophilia B program in the European Union and certain other countries.

 

Our existing collaborations, and any future collaborations we enter, may pose several risks, including the following:

 

·                   collaborators have significant discretion in determining the efforts and resources that they will apply to these collaborations;

·                   we generally have limited or no control over the design or conduct of clinical trials sponsored by our current collaborators;

·                   we may be hampered from entering into collaboration arrangements if we are unable to obtain consent form our licensor to enter into sublicensing arrangements of technology we have licensed from such licensors;

·                   if our collaborators do not conduct the clinical trials they sponsor in accordance with regulatory requirements or stated protocols, we will not be able to rely on the data produced in such trials in our further development efforts;

·                   collaborators may not perform their obligations as expected;

·                   collaborators may also have relationships with other entities, some of which may be our competitors;

·                   collaborators may not pursue development and commercialization of any product candidates or may elect not to continue or renew development or commercialization programs based on clinical trial results, changes in the collaborators’ strategic focus or available funding, or external factors, such as an acquisition, that divert resources or create competing priorities;

·                   collaborators may delay clinical trials, provide insufficient funding for a clinical trial program, stop a clinical trial or abandon a product candidate, repeat or conduct new clinical trials or require a new formulation of a product candidate for clinical testing;

·                   collaborators could develop, independently or with third parties, products that compete directly or indirectly with our products or product candidates, if the collaborators believe that competitive products are more likely to be successfully developed or can be commercialized under terms that are more economically attractive than ours;

·                   our collaboration arrangements may impose restrictions on our ability to undertake other development efforts that may appear to be attractive to us;

·                   product candidates discovered in collaboration with us may be viewed by our collaborators as competitive with their own product candidates or products, which may cause collaborators to cease to devote resources to the commercialization of our product candidates;

 

43



Table of Contents

 

·                   a collaborator with marketing and distribution rights that achieves regulatory approval may not commit sufficient resources to the marketing and distribution of such product or products;

·                   disagreements with collaborators, including over proprietary rights, contract interpretation or the preferred course of development, could cause delays or termination of the research, development or commercialization of product candidates, lead to additional responsibilities for us, delay or impede reimbursement of certain expenses or result in litigation or arbitration, any of which would be time-consuming and expensive;

·                   collaborators may not properly maintain or defend our intellectual property rights or may use our proprietary information in such a way as to invite litigation that could jeopardize or invalidate our rights or expose us to potential litigation;

·                   collaborators may infringe the intellectual property rights of third parties, which may expose us to litigation and potential liability; and

·                   collaborations may in some cases be terminated for the convenience of the collaborator and, if terminated, we could be required to expend additional funds to pursue further development or commercialization of the applicable product or product candidates.

 

If our collaborations do not result in the successful development and commercialization of products or if one of our collaborators terminates its agreement with us, we may not receive future research funding or milestone or royalty payments under the collaboration, and we may lose access to important technologies and capabilities of the collaboration. All of the risks relating to product development, regulatory approval and commercialization described herein also apply to the activities of our development collaborators.

 

Risks Related to Our Intellectual Property

 

We rely on licenses of intellectual property from third parties, and such licenses may not provide adequate rights or may not be available in the future on commercially reasonable terms or at all, and our licensors may be unable to obtain and maintain patent protection for the technology or products that we license from them.

 

We currently are heavily reliant upon licenses of proprietary technology from third parties that is important or necessary to the development of our technology and products, including technology related to our manufacturing process, our vector platform, our gene cassettes and the therapeutic genes of interest we are using. These and other licenses may not provide adequate rights to use such technology in all relevant fields of use. Licenses to additional third party technology that may be required for our development programs may not be available in the future or may not be available on commercially reasonable terms, which could have a material adverse effect on our business and financial condition.

 

In some circumstances, we may not have the right to control the preparation, filing and prosecution of patent applications, or to maintain the patents, covering technology that we license from third parties. In addition, some of our agreements with our licensors require us to obtain consent from the licensor before we can enforce patent rights, and our licensor may withhold such consent or may not provide it on a timely basis. Therefore, we cannot be certain that these patents and applications will be prosecuted and enforced in a manner consistent with the best interests of our business. In addition, if third parties who license patents to us fail to maintain such patents, or lose rights to those patents, the rights we have licensed may be reduced or eliminated.

 

Our intellectual property licenses with third parties may be subject to disagreements over contract interpretation, which could narrow the scope of our rights to the relevant intellectual property or technology or increase our financial or other obligations to our licensors.

 

The agreements under which we license intellectual property or technology from third parties are complex, and certain provisions in such agreements may be susceptible to multiple interpretations. The resolution of any contract interpretation disagreement that may arise could narrow what we believe to be the scope of our rights to the relevant intellectual property or technology, or increase what we believe to be our financial or other obligations under the relevant agreement, either of which could have a material adverse effect on our business and financial condition.

 

44



Table of Contents

 

If we fail to comply with our obligations in our intellectual property licenses with third parties, we could lose rights that are important to our business.

 

We in-license intellectual property from third parties that is material to our product candidates, including technology related to our manufacturing process, our vector platform, and the therapeutic genes and gene cassettes we are using. Our licensing arrangements with third parties impose diligence, development and commercialization timelines, milestone payment, royalty, insurance and other obligations on us. If we fail to comply with these obligations, our counterparties may have the right to terminate these agreements, in which case we might not be able to develop, manufacture or market any product that is covered by these agreements or may face other penalties under the agreements. Such an occurrence could materially adversely affect the value of the product candidate being developed under any such agreement. Termination of these agreements or reduction or elimination of our rights under these agreements may result in our having to negotiate new or amended agreements with less favorable terms, or cause us to lose our rights under these agreements, including our rights to important intellectual property or technology.

 

If we are unable to obtain and maintain patent protection for our technology and products, or if the scope of the patent protection is not sufficiently broad, our ability to successfully commercialize our products may be impaired.

 

We rely upon a combination of in-licensed and owned patents, trade secret protection and confidentiality agreements to protect our intellectual property. Our success depends in a large part on our ability to obtain and maintain this protection in the European Union, the United States and other countries, in part by filing patent applications related to our novel technologies and product candidates. Our patents may not provide us with any meaningful commercial protection, prevent competitors from competing with us or otherwise provide us with any competitive advantage. Our competitors may be able to circumvent our owned or licensed patents by developing similar or alternative technologies or products in a non-infringing manner. Successful challenges to our patents may result in loss of exclusivity or freedom to operate or in patent claims being narrowed, invalidated or held unenforceable, in whole or in part, which could limit our ability to stop others from using or commercializing similar or identical technology and products, or limit the duration of the patent protection of our technology and products.

 

The patent prosecution process is expensive, time-consuming and uncertain, and we may not be able to file and prosecute all necessary or desirable patent applications at a reasonable cost or in a timely manner. It is also possible that we will fail to identify patentable aspects of our research and development output before it is too late to obtain patent protection. Additionally, given the amount of time required for the development, testing and regulatory review of new product candidates, patents protecting such candidates might expire before or shortly after such candidates are commercialized. As a result, our owned and licensed patent portfolio may not provide us with sufficient rights to exclude others from commercializing products similar or identical to ours.

 

The patent position of biotechnology and pharmaceutical companies generally is highly uncertain, involves complex legal and factual questions and has in recent years been the subject of much litigation. In addition, the laws of foreign countries may not protect our rights to the same extent as the laws of the United States. For example, EU patent law with respect to the patentability of methods of treatment of the human body is more limited than U.S. law. Publications of discoveries in the scientific literature often lag behind the actual discoveries, and patent applications in the United States and other jurisdictions are typically not published until 18 months after their priority date, or in some cases at all. Therefore, we cannot know with certainty whether we were the first to make the inventions or that we were the first to file for patent protection of the inventions claimed in our owned or licensed patents or pending patent applications. As a result, the issuance, scope, validity, enforceability and commercial value of our patent rights are highly uncertain. Our pending and future patent applications may not result in patents being issued that protect our technology or products, in whole or in part, or which effectively prevent others from commercializing competitive technologies and products. Changes in either the patent laws or interpretation of the patent laws in the European Union, the United States or other countries may diminish the value of our patents or narrow the scope of our patent protection.

 

We may become involved in lawsuits to protect or enforce our patents or other intellectual property or third parties may assert their intellectual property rights against us, which could be expensive, time consuming and unsuccessful.

 

Competitors may infringe our owned or licensed patents or other intellectual property. To counter infringement or unauthorized use, we may be required to file infringement claims, which can be expensive and

 

45



Table of Contents

 

time consuming. An adverse result in any litigation proceeding could put one or more of our patents at risk of being invalidated, maintained in more narrowly amended form or interpreted narrowly.

 

Even if resolved in our favor, litigation or other legal proceedings relating to intellectual property claims may cause us to incur significant expenses, increase our operating losses, reduce available resources and could distract our technical and management personnel from their normal responsibilities. In addition, there could be public announcements of the results of hearings, motions or other interim proceedings or developments, which could have an adverse effect on the price of our common shares.

 

Third parties may initiate legal proceedings alleging that we are infringing their intellectual property rights, the outcome of which would be uncertain and could have a material adverse effect on the success of our business. If we are found to infringe a third party’s intellectual property rights, we could be required to obtain a license from such third party to continue developing and marketing our products and technology. We may not be able to obtain the required license on commercially reasonable terms or at all. Even if we were able to obtain a license, it could be non-exclusive, thereby giving our competitors access to the same technologies licensed to us. We could be forced, including by court order, to cease commercializing the infringing technology or product or otherwise to cease using the relevant intellectual property. In addition, we could be found liable for monetary damages, including treble damages and attorneys’ fees if we are found to have willfully infringed a patent. A finding of infringement could prevent us from commercializing our product candidates or force us to cease or materially modify some of our business operations, which could materially harm our business. Claims that we have misappropriated the confidential information or trade secrets of third parties could have a similar negative impact on our business.

 

For example, we are aware of patents owned by third parties that relate to some aspects of our programs that are still in development. In some cases, because we have not determined the final methods of manufacture, the method of administration or the therapeutic compositions for these programs, we cannot determine whether rights under such third party patents will be needed. In addition, in some cases, we believe that the claims of these patents are invalid or not infringed, or will expire before commercialization. However, if such patents are needed and found to be valid and infringed, we could be required to obtain licenses, which might not be available on commercially reasonable terms, or to cease or delay commercializing certain product candidates, or to change our programs to avoid infringement.

 

Our reliance on third parties may require us to share our trade secrets, which could increase the possibility that a competitor will discover them or that our trade secrets will be misappropriated or disclosed.

 

Because we collaborate with various organizations and academic research institutions on the advancement of our gene therapy platform, we must, at times, share trade secrets with them. We seek to protect our proprietary technology in part by entering into confidentiality agreements and, if applicable, materials transfer agreements, collaborative research agreements, consulting agreements or other similar agreements with our collaborators, advisors and consultants prior to beginning research or disclosing proprietary information. These agreements typically limit the rights of the third parties to use or disclose our confidential information, such as trade secrets. Despite the contractual provisions employed when working with third parties, the need to share trade secrets and other confidential information increases the risk that such trade secrets become known by our competitors, are inadvertently incorporated into the technology of others, or are disclosed or used in violation of these agreements. Given that our proprietary position is based, in part, on our know-how and trade secrets, a competitor’s discovery of our trade secrets or other unauthorized use or disclosure would impair our competitive position and may have a material adverse effect on our business.

 

In addition, these agreements typically restrict the ability of our collaborators, advisors and consultants to publish data potentially relating to our trade secrets. Our academic collaborators typically have rights to publish data, provided that we are notified in advance and may delay publication for a specified time in order to secure our intellectual property rights arising from the collaboration. In other cases, publication rights are controlled exclusively by us, although in some cases we may share these rights with other parties. We also conduct joint research and development programs that may require us to share trade secrets under the terms of our research and development partnerships or similar agreements.

 

Some courts inside and outside the United States are less willing or unwilling to protect trade secrets. If any of our trade secrets were to be lawfully obtained or independently developed by a competitor, we would

 

46



Table of Contents

 

have no right to prevent them, or those to whom they communicate it, from using that technology or information to compete with us.

 

47



Table of Contents

 

Risks Related to Pricing and Reimbursement

 

We face uncertainty related to insurance coverage of, and pricing and reimbursement for product candidates for which we may receive marketing approval.

 

We anticipate that the cost of treatment using our product candidates will be significant. We expect that most patients and their families will not be capable of paying for our products themselves. There will be no commercially viable market for our product candidates without reimbursement from third party payers, such as government health administration authorities, private health insurers and other organizations. Even if there is a commercially viable market, if the level of third party reimbursement is below our expectations, our revenues and gross margins will be adversely affected and our business will be harmed.

 

Government authorities and other third party payers, such as private health insurers and health maintenance organizations, decide which medications they will pay for and establish reimbursement levels. Reimbursement systems vary significantly by country and by region, and reimbursement approvals must be obtained on a country-by-country basis. Government authorities and third party payers have attempted to control costs by limiting coverage and the amount of reimbursement for particular medications and procedures. Increasingly, third party payers require drug companies to provide them with predetermined discounts from list prices, are exerting influence on decisions regarding the use of particular treatments and are limiting covered indications. Additionally, in the United States and some foreign jurisdictions, legislative and regulatory changes regarding the healthcare system and insurance coverage, particularly in light of the new U.S. presidential administration, could result in more rigorous coverage criteria and downward pressure on drug prices, and may affect our ability to profitably sell any products for which we obtain marketing approval.

 

The pricing review period and pricing negotiations for new medicines take considerable time and have uncertain results. Pricing review and negotiation usually begins only after the receipt of regulatory marketing approval, and some authorities require approval of the sale price of a product before it can be marketed. In some markets, particularly the countries of the European Union, prescription pharmaceutical pricing remains subject to continuing direct governmental control and to drug reimbursement programs even after initial approval is granted and price reductions may be imposed. Prices of medical products may also be subject to varying price control mechanisms or limitations as part of national health systems if products are considered not cost-effective or where a drug company’s profits are deemed excessive. In addition, pricing and reimbursement decisions in certain countries can lead to mandatory price reductions or additional reimbursement restrictions in other countries. As a result of these restrictions, any product candidates for which we may obtain marketing approval may be subject to price regulations that delay or prohibit our or our partners’ commercial launch of the product in a particular jurisdiction. In addition, we or our collaborators may elect to reduce the price of our products in order to increase the likelihood of obtaining reimbursement approvals. In the event that countries impose prices, which are not sufficient to allow us or our collaborators to generate a profit, we or our collaborators may refuse to launch the product in such countries or withdraw the product from the market. If pricing is set at unsatisfactory levels, or if the price decreases, our business could be harmed, possibly materially. If we fail to obtain and sustain an adequate level of coverage and reimbursement for our products by third party payers, our ability to market and sell our products would be adversely affected and our business would be harmed.

 

Due to the generally limited addressable market for our target orphan indications and the potential for our therapies to offer therapeutic benefit in a single administration, we face uncertainty related to pricing and reimbursement for these product candidates.

 

The relatively small market size for orphan indications and the potential for long-term therapeutic benefit from a single administration present particular challenges to pricing review and negotiation of our product candidates for which we may obtain marketing authorization. The patient populations of our product candidates targeted at orphan and ultra-orphan diseases are relatively small. If we are unable to obtain adequate levels of reimbursement relative to the small market size in our target orphan and ultra-orphan indications, our ability to support our development and commercial infrastructure and to successfully market and sell our product candidates for which we may obtain marketing approval will be adversely affected.

 

We also anticipate that many or all of our gene therapy product candidates may provide long-term, and potentially curative benefit, with a single administration. This is a different paradigm than that of other pharmaceutical therapies, which often require an extended course of treatment or frequent administration. As a result, governments and other payers may be reluctant to provide the significant level of reimbursement that we

 

48



Table of Contents

 

seek at the time of administration of our gene therapies or may seek to tie reimbursement to clinical evidence of continuing therapeutic benefit over time. Although we anticipate that our product candidates will need to be administered only once, there may be situations in which re-administration is required, which may further complicate the pricing and reimbursement for these treatments. In addition, in light of the anticipated cost of these therapies, governments and other payers may be particularly restrictive in making coverage decisions. These factors could limit our commercial success and harm our business.

 

Risks Related to Our Financial Position and Need for Additional Capital

 

We have incurred significant losses to date, expect to incur losses over the next several years and may never achieve or maintain profitability.*

 

We had a net loss of $20.3 million in the first quarter of 2017, $73.4 million in full year 2016 and $82.1 million in 2015. As of March 31, 2017, we had an accumulated deficit of $416.3 million. To date, we have financed our operations primarily through the sale of equity securities and convertible debt, venture loans, through upfront payments from our collaboration partners and, to a lesser extent, subsidies and grants from governmental agencies and fees for services. We have devoted substantially all of our financial resources and efforts to research and development, including preclinical studies and clinical trials. A significant portion of potential consideration under our agreement with BMS is contingent on achieving research, development, regulatory and sales milestones. We expect to continue to incur significant expenses and losses over the next several years, and our net losses may fluctuate significantly from quarter to quarter and year to year.

 

We anticipate that our expenses will increase substantially as we:

 

·                   prepare for a pivotal study for our gene therapy candidate in hemophilia B, in collaboration with Chiesi;

·                   advance the preclinical development and initiate a clinical study for our product candidate in Huntington’s disease;

·                   progress our research programs of additional product candidates targeting liver-directed and CNS disorders;

·                   conduct any additional trials or tests beyond those originally anticipated in order to confirm the safety or efficacy of our product candidates;

·                   seek marketing approval for any product candidates that successfully complete clinical trials;

·                   acquire or in-license rights to new therapeutic targets or product candidates;

·                   enter into collaboration agreements with third parties to collaborate on the research and development of potential product candidates;

·                   build clinical, medical, regulatory affairs, development and commercial infrastructure in the United States;

·                   maintain, expand and protect our intellectual property portfolio, including in-licensing additional intellectual property rights from third parties;

·                   hire additional executives to fill current vacancies, including a Chief Financial Officer and Chief Operating Officer; and

·                   incur cost to terminate or retain employees related to restructuring our operations.

 

We and our collaborators may never succeed in these activities and, even if we do, may never generate revenues that are sufficient to achieve or sustain profitability. Our failure to become and remain profitable would depress the value of our company and could impair our ability to expand our business, maintain our research and development efforts, diversify our product offerings or even continue our operations.

 

We will likely need to raise additional funding, which may not be available on acceptable terms, or at all. Failure to obtain capital when needed may force us to delay, limit or terminate our product development efforts or other operations which could have a material adverse effect on our business, financial conditions, results of operations and cash flows.

 

We expect to incur significant expenses in connection with our on-going activities and that we will likely need to obtain substantial additional funding in connection with our continuing operations. In addition, we have based our estimate of our financing requirements on assumptions that may prove to be wrong, and we could use our capital resources sooner than we currently expect.

 

49



Table of Contents

 

Adequate capital may not be available to us when needed or may not be available on acceptable terms. Our ability to obtain debt financing may be limited by covenants we have made under our Loan and Security Agreement with Hercules Technology Growth Capital, Inc. (“Hercules”) and our pledge to Hercules of substantially all of our assets as collateral. If we raise additional capital through the sale of equity or convertible debt securities, our shareholders’ ownership interest will be diluted, and the terms of these securities may include liquidation or other preferences that adversely affect the rights of holders of our common shares.

 

If we raise additional funds through collaborations, strategic alliances, or marketing, distribution or licensing arrangements with third parties, we may have to issue additional equity, relinquish valuable rights to our technologies, future revenue streams, products or product candidates, or grant licenses on terms that may not be favorable to us. If we are unable to raise capital when needed or on attractive terms, we could be forced to delay, reduce or eliminate our research and development programs or any future commercialization efforts, which would have a negative impact on our financial condition, results of operations and cash flows. If our collaborations with BMS and Chiesi are not successful, our development plans, financial position and opportunities for growth may be adversely affected.

 

Our existing and any future indebtedness could adversely affect our ability to operate our business.*

 

As of March 31, 2017, we had $20.4 million of outstanding borrowings under our Loan and Security Agreement with Hercules, which we are required to repay in monthly principal installments from December 2017 through May 2020. We could in the future incur additional debt obligations beyond our borrowings from Hercules. Our existing loan obligations, together with other similar obligations that we may incur in the future, could have significant adverse consequences, including:

 

·                       requiring us to dedicate a portion of our cash resources to the payment of interest and principal, reducing money available to fund working capital, capital expenditures, research and development and other general corporate purposes;

·                       increasing our vulnerability to adverse changes in general economic, industry and market conditions;

·                       subjecting us to restrictive covenants that may reduce our ability to take certain corporate actions or obtain further debt or equity financing;

·                       limiting our flexibility in planning for, or reacting to, changes in our business and the industry in which we compete; and

·                       placing us at a disadvantage compared to our competitors that have less debt or better debt servicing options.

 

We may not have sufficient funds, and may be unable to arrange for additional financing, to pay the amounts due under our existing loan obligations. Failure to make payments or comply with other covenants under our existing debt could result in an event of default and acceleration of amounts due. Under our agreement with Hercules, the occurrence of an event that would reasonably be expected to have a material adverse effect on our business, operations, assets or condition is an event of default. If an event of default occurs and the lender accelerates the amounts due, we may not be able to make accelerated payments, and the lender could seek to enforce security interests in the collateral securing such indebtedness, which includes substantially all of our assets.

 

We may be required to sublease space in excess of our requirements at our Amsterdam site.*

 

In March 2016, we entered into a lease for a new approximately 100,000 square feet facility in Amsterdam, and we amended this agreement in June 2016 in order to lease an additional 11,000 square feet. We intend to initiate the consolidation of our current three Amsterdam sites into the new site in the first half of 2017. The lease for this facility terminates in 2032. Following our restructuring announced in November 2016, we do not expect to utilize a significant portion of our new Amsterdam facility. We are contractually required to incur significant costs in relation to areas not utilized by us over the full contractual term. While we will seek to sublease any excess space, we may not be able to do so at commercially attractive terms.

 

50



Table of Contents

 

Risks Related to Other Legal Compliance Matters

 

Our relationships with customers and third party payers will be subject to applicable anti-kickback, anti-bribery, fraud and abuse and other laws and regulations, which could expose us to criminal sanctions, civil penalties, contractual damages, reputational harm and diminished profits and future earnings.

 

Healthcare providers, physicians and third party payers will play a primary role in the recommendation and prescription of any products for which we obtain marketing approval. Our future arrangements with third party payers and customers may expose us to broadly applicable anti-bribery laws, including the Foreign Corrupt Practices Act, as well as fraud and abuse and other healthcare laws and regulations that may constrain the business or financial arrangements and relationships through which we market, sell and distribute any products for which we obtain marketing approval.

 

Efforts to ensure that our business arrangements with third parties will comply with applicable laws and regulations will involve substantial costs. If our operations, or the activities of our collaborators, distributors or other third party agents are found to be in violation of any of these laws or any other governmental regulations that may apply to us, we may be subject to significant civil, criminal and administrative penalties, damages, fines, imprisonment, exclusion of products from government funded healthcare programs and the curtailment or restructuring of our operations.

 

If we fail to comply with environmental, health and safety laws and regulations, we could become subject to fines or penalties or incur costs that could harm our business.

 

We are subject to numerous environmental, health and safety laws and regulations, including those governing laboratory procedures and the handling, use, storage, treatment and disposal of hazardous materials and wastes. Our operations involve the use of hazardous and flammable materials, including chemicals and biological materials. Our operations also produce hazardous waste products. We cannot eliminate the risk of contamination or injury from these materials. We also could incur significant costs associated with civil or criminal fines and penalties for failure to comply with such laws and regulations.

 

Although we maintain employer’s liability insurance to cover us for costs and expenses we may incur due to injuries to our employees resulting from the use of hazardous materials, this insurance may not provide adequate coverage against potential liabilities. We do not maintain insurance for environmental liability or toxic tort claims that may be asserted against us in connection with our storage or disposal of biological, hazardous or radioactive materials.

 

In addition, we may incur substantial costs in order to comply with current or future environmental, health and safety laws and regulations. These current or future laws and regulations may impair our research, development or production efforts. Our failure to comply with these laws and regulations also may result in substantial fines, penalties or other sanctions.

 

Product liability lawsuits could cause us to incur substantial liabilities and to limit commercialization of our therapies.

 

We face an inherent risk of product liability related to the testing of our product candidates in human clinical trials and in connection with product sales. If we cannot successfully defend ourselves against claims that our product candidates or products caused injuries, we will incur substantial liabilities. Regardless of merit or eventual outcome, liability claims may result in:

 

·                   decreased demand for any product candidates or products that we develop or sell;

·                   injury to our reputation and significant negative media attention;

·                   negative publicity or public opinion surrounding gene therapy;

·                   withdrawal of clinical trial participants;

·                   significant costs to defend the related litigation;

·                   substantial monetary awards to trial participants or patients;

·                   loss of revenue;

·                   reduced resources of our management to pursue our business strategy; and

·                   the inability to further develop or commercialize any products that we develop.

 

51



Table of Contents

 

Dependent upon the country where the clinical trial is conducted, we currently hold a maximum of €6,000,000 and minimum of €2,000,000 in clinical trial insurance coverage in the aggregate, with a per incident limit of €450,000 to €1,000,000 with respect to the clinical studies we conduct. Such coverage may not be adequate to cover all liabilities that we may incur. We may need to increase our insurance coverage as we expand our clinical trials. In addition, insurance coverage is increasingly expensive. We may not be able to maintain insurance coverage at a reasonable cost or in an amount adequate to satisfy any liability that may arise.

 

Risks Related to Employee Matters and Managing Growth

 

Our future success depends on our ability to retain key executives and technical staff and to attract, retain and motivate qualified personnel.

 

We are highly dependent on the research and development, clinical and business development expertise of our Chief Executive Officer, Mathew Kapusta; our Chief Medical Officer, Christian Meyer, M.D. and our Chief Scientific Officer, Harald Petry as well as the other principal members of our management, scientific and clinical team. Although we have entered into employment agreements with our senior management, each of them may terminate their employment on relatively short notice. We do not maintain key person insurance for any of our senior management or employees.

 

The loss of the services of our senior management or other key employees could impede the achievement of our research, development and commercialization objectives and seriously harm our ability to successfully implement our business strategy. Furthermore, replacing senior management and key employees may be difficult and may take an extended period of time because of the limited number of individuals in our industry with the breadth and depth of skills and experience required to successfully develop, gain regulatory approval of and commercialize gene therapy products. Competition to hire from this limited pool is intense, and we may be unable to hire, train, retain or motivate these key personnel on acceptable terms.

 

If we are unable to continue to attract and retain high quality personnel, our ability to pursue our growth strategy will be limited.

 

We have initiated a significant restructuring of our operations that will include a sizable reduction in headcount in our Amsterdam facility and expansion of personnel in our Lexington facility, and as a result, we may encounter challenges managing the associated organizational change.*

 

In November 2016, we announced plans to restructure and refocus our business, which will result in the elimination of approximately 50 to 55 positions, as well as the transition of certain operations to our Lexington, Massachusetts facility. We will incur expenses related to the reduction in staff as well as the retention of key personnel. We may also experience operational disruptions as we implement our new organizational structure and transfer certain functions to Lexington. This process may distract the attention of management and staff and may cause disruption in our operations.

 

At the same time, we continue to expand the scope of certain of our operations in the United States, particularly in the areas of clinical operations, medical and regulatory affairs and product development. To manage our operations and new organizational structure, we will be required to implement and improve our managerial, operational and financial systems and recruit and train additional qualified personnel. Due to our limited financial resources and the limited experience of our management team in implementing significant organizational change, we may not be able to effectively manage this process.

 

Risks Related to Our Common Shares

 

The price of our common shares has been and may in the future be volatile and fluctuate substantially.*

 

Our share price has been and may in the future be volatile. From the start of trading of our common shares on the NASDAQ Global Select Market on February 4, 2014 through May 1, 2017, the sale price of our common shares ranged from a high of $36.38 to a low of $4.91. The closing price on May 1, 2017, was $5.46 per common share. The stock market in general and the market for smaller biopharmaceutical companies in particular have experienced extreme volatility that has often been unrelated to the operating performance of particular companies. The market price for our common shares may be influenced by many factors, including:

 

·                   the success of competitive products or technologies;

·                   results of clinical trials of our product candidates or those of our competitors;

 

52



Table of Contents

 

·                   public perception of gene therapy;

·                   regulatory delays and greater government regulation of potential products due to adverse events;

·                   regulatory or legal developments in the European Union, the United States and other countries;

·                   developments or disputes concerning patent applications, issued patents or other proprietary rights;

·                   the recruitment or departure of key personnel;

·                   the level of expenses related to any of our product candidates or clinical development programs;

·                   the results of our efforts to discover, develop, acquire or in- license additional product candidates or products;

·                   actual or anticipated changes in estimates as to financial results, development timelines or recommendations by securities analysts;

·                   variations in our financial results or those of companies that are perceived to be similar to us;

·                   changes in the structure of healthcare payment systems;

·                   market conditions in the pharmaceutical and biotechnology sectors; and

·                   general economic, industry and market conditions.

 

An active trading market for our common shares may not be sustained.

 

Although our common shares are listed on The NASDAQ Global Select Market, an active trading market for our shares may not be sustained. If an active market for our common shares does not continue, it may be difficult for our shareholders to sell their shares without depressing the market price for the shares or sell their shares at all. Any inactive trading market for our common shares may also impair our ability to raise capital to continue to fund our operations by selling shares and may impair our ability to acquire other companies or technologies by using our shares as consideration.

 

Our directors and named executive officers and major shareholders, if they choose to act together, will continue to have a significant degree of control with respect to matters submitted to shareholders for approval. *

 

Our directors and named executive offices and major shareholders more than 5% of our outstanding common shares, in the aggregate, beneficially own approximately 31.5% of our issued shares as at March 31, 2017. As a result, if these shareholders were to choose to act together, they may be able, as a practical matter, to control many matters submitted to our shareholders for approval, as well as our management and affairs. For example, these persons, if they choose to act together, could control the election of board directors and approval of any merger, consolidation or sale of all or substantially all of our assets.

 

Provisions of our articles of association or Dutch corporate law might deter acquisition bids for us that might be considered favorable and prevent or frustrate any attempt to replace our board.

 

Certain provisions of our articles of association may make it more difficult for a third party to acquire control of us or effect a change in our board. These provisions include:

 

·                   staggered terms of our directors;

·                   a provision that our directors may only be removed at a general meeting of shareholders by a two-thirds majority of votes cast representing more than half of the issued share capital of the company (unless the removal was proposed by the board); and

·                   a requirement that certain matters, including an amendment of our articles of association, may only be brought to our shareholders for a vote upon a proposal by our board.

 

We do not expect to pay dividends in the foreseeable future.

 

We have not paid any dividends since our incorporation. Even if future operations lead to significant levels of distributable profits, we currently intend that earnings, if any, will be reinvested in our business and that dividends will not be paid until we have an established revenue stream to support continuing dividends. Accordingly, shareholders cannot rely on dividend income from our common shares and any returns on an investment in our common shares will likely depend entirely upon any future appreciation in the price of our common shares.

 

53



Table of Contents

 

We are an “emerging growth company,” and the reduced disclosure requirements applicable to emerging growth companies may make our common shares less attractive to investors.

 

We are an emerging growth company, as defined in the Jumpstart Our Business Startups Act of 2012 (“JOBS Act”) and may remain an emerging growth company for up to five years from our initial public offering. For so long as we remain an emerging growth company, we are permitted and intend to rely on exemptions from certain disclosure requirements that are applicable to other public companies that are not emerging growth companies. These exemptions include:

 

·                   not being required to comply with the auditor attestation requirements in the assessment of our internal control over financial reporting; and

·                   not being required to comply with any requirement that may be adopted by the Public Company Accounting Oversight Board regarding mandatory audit firm rotation or a supplement to the auditor’s report providing additional information about the audit and the financial statements.

 

If some investors find our common shares less attractive as a result of our reliance on these exemptions, trading market for our common shares may be less active and our share price may be more volatile.

 

We ceased to qualify as a foreign private issuer as of January 1, 2017, and therefore must comply with the Exchange Act, which will result in additional legal, accounting and other expenses.

 

Beginning in January 2017, we must comply with the Exchange Act reporting and other requirements applicable to U.S. domestic filers, which are more detailed and extensive than the requirements for foreign private issuers to which we were previously subject. In addition, we are now required to report our financial results under U.S. GAAP, including our historical financial results, which have previously been prepared in accordance with International Financial Reporting Standards (“IFRS”). We have made changes in our corporate governance practices in accordance with various SEC and NASDAQ rules. The transition to U.S. GAAP reporting has required additional expenditures, and the related regulatory, compliance and insurance costs to us may be significantly higher than the costs we incurred as a foreign private issuer.

 

If we fail to implement and maintain an effective system of internal controls, we may be unable to accurately report our results of operations or prevent fraud or fail to meet our reporting obligations, and investor confidence and the market price of our common shares may be materially and adversely affected.

 

If we fail to achieve and maintain the adequacy of our internal control over financial reporting, we may not be able to conclude on an ongoing basis that we have effective internal control over financial reporting. If we fail to achieve and maintain effective internal control over financial reporting, we could experience material misstatements in our financial statements and fail to meet our reporting obligations, which would likely cause investors to lose confidence in our reported financial information. This could in turn limit our access to capital markets, harm our results of operations, and lead to a decline in the trading price of our common shares. Additionally, ineffective internal control over financial reporting could expose us to increased risk of fraud or misuse of corporate assets and subject us to potential delisting from The NASDAQ Global Select Market, regulatory investigations and civil or criminal sanctions. Our reporting and compliance obligations may place a significant strain on our management, operational and financial resources and systems for the foreseeable future. We have previously identified material weaknesses in our internal controls, and our remediation efforts in this regard may not be effective.

 

Risks for U.S. Holders

 

We qualify as a passive foreign investment company as of December 31, 2016, which may result in adverse U.S. federal income tax consequence to U.S. holders.

 

Based on our average value of our gross assets, our cash and cash equivalents as well as the price of our shares we qualify as a passive foreign investment company (“PFIC”) for U.S. federal income tax for 2016. A corporation organized outside the United States generally will be classified as a PFIC for U.S. federal income tax purposes in any taxable year in which at least 75% of its gross income is passive income or on average at least 50% of the gross value of its assets is attributable to assets that produce passive income or are held for the production of passive income. Passive income for this purpose generally includes dividends, interest, royalties, rents and gains from commodities and securities transactions. Our status in any taxable year will depend on our assets and activities in each year, and because this is a factual determination made annually after the end of each taxable year, there can be no assurance that we will continue to qualify as a PFIC in future taxable years. The market value of our assets may be determined in large part by reference to the market price of our common shares, which is likely to fluctuate, and may fluctuate considerably given that market prices of biotechnology

 

54



Table of Contents

 

companies have been especially volatile. As we are a PFIC for the taxable year 2016, certain adverse U.S. federal income tax consequences, including reporting obligations, could apply to a U.S. holder who held our common shares during 2016.

 

Any U.S. or other foreign judgments may be difficult to enforce against us in the Netherlands.

 

Although we now report as a U.S. domestic filer for SEC reporting purposes, we are incorporated under the laws of the Netherlands. Some of the members of our board and senior management reside outside the United States. As a result, it may not be possible for shareholders to effect service of process within the United States upon such persons or to enforce judgments against them or us in U.S. courts, including judgments predicated upon the civil liability provisions of the federal securities laws of the United States. In addition, it is not clear whether a Dutch court would impose civil liability on us or any of our managing directors or supervisory directors in an original action based solely upon the federal securities laws of the United States brought in a court of competent jurisdiction in the Netherlands.

 

The United States and the Netherlands currently do not have a treaty providing for the reciprocal recognition and enforcement of judgments, other than arbitration awards, in civil and commercial matters. Consequently, a final judgment for payment given by a court in the United States, whether or not predicated solely upon U.S. securities laws, would not automatically be recognized or enforceable in the Netherlands. In order to obtain a judgment which is enforceable in the Netherlands, the party in whose favor a final and conclusive judgment of the U.S. court has been rendered will be required to file its claim with a court of competent jurisdiction in the Netherlands. Such party may submit to the Dutch court the final judgment rendered by the U.S. court. If and to the extent that the Dutch court finds that the jurisdiction of the U.S. court has been based on grounds which are internationally acceptable and that proper legal procedures have been observed, the Dutch court will, in principle, give binding effect to the judgment of the U.S. court, unless such judgment contravenes principles of public policy of the Netherlands. Dutch courts may deny the recognition and enforcement of punitive damages or other awards. Moreover, a Dutch court may reduce the amount of damages granted by a U.S. court and recognize damages only to the extent that they are necessary to compensate actual losses or damages. Enforcement and recognition of judgments of U.S. courts in the Netherlands are solely governed by the provisions of the Dutch Civil Procedure Code.

 

Therefore U.S. shareholders may not be able to enforce against us or our board members or senior management who are residents of the Netherlands or countries other than the United States any judgments obtained in U.S. courts in civil and commercial matters, including judgments under the U.S. federal securities laws.

 

The rights and responsibilities of our shareholders and directors are governed by Dutch law and differ in some important respects from the rights and responsibilities of shareholders under U.S. law.

 

Although we now report as a U.S. domestic filer for SEC purposes, our corporate affairs are governed by our articles of association and by the laws governing companies incorporated in the Netherlands. The rights of our shareholders and the responsibilities of members of our board under Dutch law are different than under the laws of some U.S. jurisdictions. In the performance of their duties, our board members are required by Dutch law to consider the interests of uniQure, its shareholders, its employees and other stakeholders and not only those of our shareholders.

 

Item 2. Unregistered Sales of Equity Securities and Use of Proceeds

 

None.

 

Item 3. Defaults Upon Senior Securities

 

None.

 

Item 4. Mine Safety Disclosures

 

None.

 

Item 5. Other Information

 

None.

 

55



Table of Contents

 

Item 6. Exhibits

 

See the Exhibit Index following the signature page to this Quarterly Report on Form 10-Q for a list of exhibits filed or furnished with this report, which Exhibit Index is incorporated herein by reference.

 

SIGNATURE

 

Pursuant to the requirements of the Securities Exchange Act of 1934, as amended, the registrant has duly caused this report to be signed on its behalf by the undersigned thereunto duly authorized.

 

 

UNIQURE, N.V.

 

 

 

 

 

By:

/s/ Matthew Kapusta

 

Matthew Kapusta

 

Chief Executive Officer and Interim Chief Financial Officer (Principal Executive Officer and Principal Financial Officer)

 

Date: May 9, 2017

 

56



Table of Contents

 

EXHIBIT INDEX

 

3.1                                Amended Articles of Association (incorporated by reference to Exhibit 1.1 of the Company’s annual report on Form 10-K for the year ended December 31, 2016 (file no. 0001-36294) filed with the Securities and Exchange Commission).

 

10.1*                  Patent License Agreement (L 107 2007), effective as of May 2, 2007, by and between the Company and the National Institutes of Health, as amended on December 31, 2009, May 31, 2013 and November 11, 2013 (a redacted version was previously filed as Exhibit 10.1 of the Company’s registration statement on form F-1 (file no. 333 193158) filed with the SEC).

 

10.2*                  Patent License Agreement (L 116 2011), effective as of August 10, 2011, by and between the Company and National Institutes of Health, as amended on May 31, 2013 and November 11, 2013 (a redacted version was previously filed as Exhibit 10.2 of the Company’s registration statement on form F-1 (file no. 333 193158) filed with the SEC).

 

10.3*                  Exclusive License Agreement, effective as of July 7, 2008, by and between the Company and St. Jude Children’s Research Hospital, Inc., as amended on July 12, 2012 (a redacted version was previously filed as Exhibit 10.10 of the Company’s registration statement on form F 1 (file no. 333 193158) filed with the SEC).

 

10.4*                  Co Development and License Agreement, entered into as of April 29, 2013, by and between the Company and Chiesi Farmaceutici S.p.A. (a redacted version was previously filed as Exhibit 10.11 of the Company’s registration statement on form F 1 (file no. 333 193158) filed with the SEC).

 

31.1*                  Rule 13a-14(a)/15d-14(a) Certification of Chief Executive Officer

 

31.2*                  Rule 13a-14(a)/15d-14(a) Certification of Chief Executive Officer

 

32.1*                  Section 1350 Certification (furnished herewith)

 

101*                     The following financial information from our Quarterly Report on Form 10-Q for the period ended March 31, 2017, filed with the Securities and Exchange Commission on May 9, 2017 is formatted in Extensible Business Reporting Language (“XBRL”): (i) Consolidated Balance Sheets; (ii) Consolidated Statements of Operations and Comprehensive Loss; (iii) Consolidated Statements of Shareholders’ Equity; (iv) Consolidated Statements of Cash Flows; and (v) Notes to Consolidated Financial Statements (tagged as blocks of text)

 


*                                          Filed herewith.

 

57


Exhibit 10.1

 

PUBLIC HEALTH SERVICE

 

PATENT LICENSE AGREEMENT - NONEXCLUSIVE

 

COVER PAGE

 

For PHS internal use only:

License Number:

L - 1 0 7 - 2007 / 0

 

License Application Number: A-274-2006

 

Serial Number(s) of Licensed Patent(s) or Patent Application(s):

 

I.                 U.S. Patent Application(s) or Patent(s):

 

(a)                        U.S. Patent Application Serial No. 09/986,618, filed November 9, 2001, now abandoned, entitled “Production of Adeno-Associated Virus in Insect Cells” [HHS Ref. No. E-325-2001/0-US-01”];

 

(b)                        U.S. Patent No. 6,723,551, issued April 20, 2004, entitled “Production of Adeno-Associated Virus in Insect Cells” [HHS Ref. No. E-325-2001/1-US-01]; and

 

(c)                         U.S. Patent Application Serial No. 10/415,834, filed May 2, 2003, entitled “Production of Adeno-Associated Virus in Insect Cells” [HHS Ref. No. E-325-2001/2-US-02].

 

II.            PCT or Foreign Patent Application(s) or Patent(s):

 

(a)                        PCT Patent Application Serial No. PCT/US02/35829, filed November 8, 2002, now abandoned, entitled “Production of Adeno-Associated Virus in Insect Cells” [HHS Ref. No. E-325-2001/2-PCT-01];

 

(b)                        Canadian Patent Application Serial No. 2,467,959, filed May 6, 2004, entitled “Production of Adeno-Associated Virus in Insect Cells” [HHS Ref. No. E-325-2001/2-CA-03];

 

(c)                         European Patent Application Serial No. 02795604.4, filed June 8, 2004, entitled “Production of Adeno-Associated Virus in Insect Cells” [HHS Ref. No. E-325-2001/2-EP-04]; and

 

(d)                        Australian Patent Application Serial No. 2002360355, filed April 28, 2004, entitled “Production of Adeno-Associated Virus in Insect Cells” [HHS Ref. No. E-325-2001/2-AU-05].

 

Licensee: Amsterdam Molecular Therapeutics

 

Cooperative Research and Development Agreement (CRADA) Number (if a subject invention): N/A

 

Additional Remarks: None

 



 

Public Benefit(s): Commercialization of this technology will benefit the public health by increasing the number of therapeutics available for the public.

 

This Patent License Agreement, hereinafter referred to as the “ Agreement ”, consists of this Cover Page, an attached Agreement , a Signature Page, Appendix A (List of Patent(s) or Patent Application(s)), Appendix B (Fields of Use and Territory), Appendix C (Royalties), Appendix D ((Benchmarks and Performance), Appendix E (Commercial Development Plan), Appendix F (Example Royalty Report), and Appendix G (Royalty Payment Options) The Parties to this Agreement are:

 

1)                                      The National Institutes of Health (“ NIH ”) or the Food and Drug Administration (“ FDA ”), hereinafter singly or collectively referred to as “ PHS ”, agencies of the United States Public Health Service within the Department of Health and Human Services (“ HHS ”); and

 

2)                                      The person, corporation, or institution identified above and on the Signature Page, having offices at the address indicated on the Signature Page, hereinafter referred to as “ Licensee .”

 



 

PHS PATENT LICENSE AGREEMENT- NONEXCLUSIVE

 

PHS and Licensee agree as follows:

 

1.                                       BACKGROUND

 

1.1                                In the course of conducting biomedical and behavioral research, PHS investigators made inventions that may have commercial applicability.

 

1.2                                By assignment of rights from PHS employees and other inventors, HHS , on behalf of the Government , owns intellectual property rights claimed in any United States or foreign patent applications or patents corresponding to the assigned inventions .  HHS also owns any tangible embodiments of these inventions actually reduced to practice by PHS .

 

1.3                                The Secretary of HHS has delegated to PHS the authority to enter into this Agreement for the licensing of rights to these inventions under 35 U.S.C. §§200-212, the Federal Technology Transfer Act of 1986, 15 U.S.C. §3710(a), and the regulations governing the licensing of Government-owned inventions, 37 CFR Part 404.

 

1.4                                PHS desires to transfer these inventions to the private sector through commercialization licenses to facilitate the commercial development of products and processes for public use and benefit.

 

1.5                                Licensee desires to acquire commercialization rights to certain of these inventions in order to develop processes, methods, or marketable products for public use and benefit.

 

2.                                       DEFINITIONS

 

2.1                                Benchmarks ” mean the performance milestones that are set forth in Appendix D.

 

2.2                                Commercial Development Plan ” means the written commercialization plan attached as Appendix E.

 

2.3                                First Commercial Sale ” means the initial transfer by or on behalf of Licensee or its sublicensees of Licensed Products or New Products by or on behalf of Licensee or its sublicensees in exchange for cash or some equivalent to which value can be assigned for the purpose of determining Net Sales .

 

2.4                                Government ” means the Government of the United States of America.

 

2.5                                Licensed Fields of Use ” means the fields of use identified in Appendix B.

 

2.6                                Licensed Patent Rights ” shall mean:

 

(a)                                  Patent applications (including provisional patent applications and PCT patent applications) or patents listed in Appendix A, all divisions and

 



 

continuations of these applications, all patents issuing from these applications, divisions, and continuations, and any reissues, reexaminations, and extensions of all these patents;

 

(b)                                  to the extent that the following contain one or more claims directed to the invention or inventions disclosed in 2.6(a):

 

(i)                                      continuations-in-part of 2.6(a);

 

(ii)                                   all divisions and continuations of these continuations-in-part;

 

(iii)                                all patents issuing from these continuations-in-part, divisions, and continuations;

 

(iv)                               priority patent application(s) of 2.6(a); and

 

(v)                                  any reissues, reexaminations, and extensions of all these patents;

 

(c)                                   to the extent that the following contain one or more claims directed to the invention or inventions disclosed in 2.6(a): all counterpart foreign and U.S. patent applications and patents to 2.6(a) and 2.6(b), including those listed in Appendix A; and

 

(d)                                  Licensed Patent Rights shall not include 2.6(b) or 2.6(c) to the extent that they contain one or more claims directed to new matter which is not the subject matter disclosed in 2.6(a).

 

2.7                                Licensed Processes ” means processes, which in the course of being practiced, would be within the scope of one or more claims of the Licensed Patent Rights that have not been held unpatentable, invalid or unenforceable by an unappealed or unappealable judgment of a court of competent jurisdiction.

 

2.8                                Licensed Products ” means (a)  Supplied Materials and (b) tangible materials, which in the course of manufacture, use, sale, or importation, would be within the scope of one or more claims of the Licensed Patent Rights that have not been held unpatentable, invalid or unenforceable by an unappealed or unappealable judgment of a court of competent jurisdiction.

 

2.9                                Licensed Territory ” means the geographical area identified in Appendix B.

 

2.10                         Net Sales ” means the total gross receipts for sales of Licensed Products or New Products by or on behalf of Licensee or its sublicensees, and from leasing, renting, or otherwise making Licensed Products or New Products available to others without sale or other dispositions, whether invoiced or not, less returns and allowances, packing costs, insurance costs, freight out, taxes or excise duties imposed on the transaction (if separately invoiced), and wholesaler and cash discounts in amounts customary in the trade to the extent actually granted No deductions shall be made for commissions paid to individuals, whether they are

 



 

with independent sales agencies or regularly employed by Licensee or its sublicensees, and on its payroll, or for the cost of collections.

 

2.11                         New Product ” means a product made using a Licensed Process but excluding Licensed Products .

 

2.12                         Practical Application ” means to manufacture in the case of a composition or product, to practice in the case of a process or method, or to operate in the case of a machine or system; and in each case, under these conditions as to establish that the invention is being utilized and that its benefits are to the extent permitted by law or Government regulations available to the public on reasonable terms.

 

2.13                         Supplied Materials ” means (a) the plasmid pFBDLSR: a plasmid for the expression of the Rep52 ORF from the baculovirus Polh promoter and the Rep78 ORF from the modified IE-1 promoter The dual Rep ORF containing plasmid is constructed by separately cloning the Rep78 and Rep52 ORF’s and combining the two ORF’s in a single expression vector/plasmid [see page 22, Paragraphs 79-81 of U.S. Patent Application Serial No. 09/986,618 (HHS Ref. No. E-325-2001/0-US-01)]; (b) the plasmid pFBAAV1pm11: a plasmid for the expression of the products of the AAV1 CAP gene which encodes the VP1, VP2 and VP3 capsid proteins from the baculovirus polh The VP genes have been modified from the wild type to produce VP1 at a level equal to VP2 without influencing VP3 expression [see pages 22-23, Paragraphs 84-85 of United States Patent Application Serial No. 09/986,618 (HHS Ref No. E-325-2001/0-US-01]; and (c) the plasmid pFBGFPR: a plasmid for the expression of GFP (green fluorescent protein) from a CMV or p10 promoter, where the promoters and GFP sequence are contained between a pair of AAV2 ITR’s Note that the plasmid has an additional cloning site which facilitates swapping-in/out eGFP with a gene of interest [see Example 1, page 20, paragraph 74 of United States Patent Application Serial No. 09/986,618 (HHS Ref. No. E-325-2001/0-US-01)] Further, these Supplied Materials were supplied by PHS to Licensee under a Non-Exclusive Patent License Agreement for Internal Commercial Use (L-043-2003/0) which was effective on May 14, 2003.

 

3.                                       GRANT OF RIGHTS

 

3.1                                PHS hereby grants and Licensee accepts, subject to the terms and conditions of this Agreement, a nonexclusive license under the Licensed Patent Rights in the Licensed Territory to make and have made, to use and have used, to sell and have sold, to offer to sell, and to import any Licensed Products or New Products in the Licensed Fields of Use , to practice and have practiced any Licensed Processes in the Licensed Fields of Use , to make, have made, to use and have used but not to sell any Supplied Materials.  As used in this Agreement , “have made” and “have used” means that Licensee shall have the limited right to use a

 



 

third party contract manufacturer to make and use only (but not to sell) Supplied Materials , Licensed Products or New Products.  Licensee acknowledges and agrees that any such third party contract manufacturer shall be bound to the terms and obligations of this Agreement .

 

3.2                                This Agreement confers no license or rights by implication, estoppel, or otherwise under any patent applications or patents of PHS other than the Licensed Patent Rights regardless of whether these patents are dominant or subordinate to the Licensed Patent Rights .

 

4.                                       SUBLICENSING

 

4.1                                Upon written approval, which shall include prior review of any sublicense agreement by PHS and which shall not be unreasonably withheld, Licensee may enter into sublicensing agreements under the Licensed Patent Rights , except that Licensee shall not have the right to solely sublicense Licensed Patent Rights.  For the avoidance of doubt .  Licensee shall only sublicense the Licensed Patent Rights in conjunction with other intellectual property owned by the Licensee or in-licensed by the Licensee .

 

4.2                                Licensee agrees that any sublicenses granted by it shall provide that the obligations to PHS of Paragraphs 5.1, 5.2, 8.1, 10.1, 10.2, 12.5 and 13.6-13.8 of this Agreement shall be binding upon the sublicensee as if it were a party to this Agreement.  Licensee further agrees to attach copies of these Paragraphs to all sublicense agreements.

 

4.3                                Any sublicenses granted by Licensee shall provide for the termination of the sublicense, or the conversion to a license directly between the sublicensee and PHS , at the option of the sublicensee, upon termination of this Agreement under Article 13 This conversion is subject to PHS approval and contingent upon acceptance by the sublicensee of the remaining provisions of this Agreement .

 

4.4                                Licensee agrees to forward PHS a complete copy of each fully executed sublicense agreement postmarked within thirty (30) days of the execution of the agreement To the extent permitted by law, PHS agrees to maintain each sublicense agreement in confidence.

 

5.                                       STATUTORY AND PHS REQUIREMENTS AND RESERVED GOVERNMENT RIGHTS

 

5.1                                Prior to the First Commercial Sale , Licensee agrees to provide PHS with reasonable quantities of Licensed Products or New Products made through the Licensed Processes or Supplied Materials solely for PHS research use, if requested in writing.

 



 

5.2                                Licensee agrees that products used or sold in the United States embodying Licensed Products or New Products or produced through use of Licensed Processes shall be manufactured substantially in the United States, unless a written waiver is obtained in advance from PHS .

 

6.                                       ROYALTIES AND REIMBURSEMENT

 

6.1                                Licensee agrees to pay PHS a noncreditable, nonrefundable license issue royalty as set forth in Appendix C.

 

6.2                                Licensee agrees to pay PHS a nonrefundable minimum annual royalty as set forth in Appendix C.

 

6.3                                Licensee agrees to pay PHS earned royalties as set forth in Appendix C.

 

6.4                                Licensee agrees to pay PHS sublicensing royalties as set forth in Appendix C.

 

6.5                                A patent or patent application licensed under this Agreement shall cease to fall within the Licensed Patent Rights for the purpose of computing earned royalty payments in any given country on the earliest of the dates that:

 

(a)                                  the application has been abandoned and not continued;

 

(b)                                  the patent expires or irrevocably lapses; or

 

(c)                                   the claim has been held to be invalid or unenforceable by an unappealed or unappealable decision of a court of competent jurisdiction or administrative agency.

 

6.6                                No multiple royalties shall be payable because any Licensed Products or New Products or Licensed Processes are covered by more than one of the Licensed Patent Rights .

 

6.7                                On sales of Licensed Products or New Products by Licensee to sublicensees or on sales made in other than an arms-length transaction, the value of the Net Sales attributed under this Article 6 to this transaction shall be that which would have been received in an arms-length transaction, based on sales of like quantity and quality products on or about the time of this transaction.

 

7.                                       PATENT FILING, PROSECUTION, AND MAINTENANCE

 

7.1                                PHS agrees to take responsibility for the preparation, filing, prosecution, and maintenance of any and all patent applications or patents included in the Licensed Patent Rights .

 

8.                                       RECORD KEEPING

 

8.1                                Licensee agrees to keep accurate and correct records of Licensed Products or New Products made, used, sold, or imported and Licensed Processes practiced under this Agreement appropriate to determine the amount of royalties due PHS.

 



 

These records shall be retained for at least five (5) years following a given reporting period and shall be available during normal business hours for inspection, at the expense of PHS , by an accountant or other designated auditor selected by PHS for the sole purpose of verifying reports and royalty payments hereunder.  The accountant or auditor shall only disclose to PHS information relating to the accuracy of reports and royalty payments made under this Agreement.  If an inspection shows an underreporting or underpayment in excess of five percent (5%) for any twelve (12) month period, then Licensee shall reimburse PHS for the cost of the inspection at the time Licensee pays the unreported royalties, including any additional royalties as required by Paragraph 9.8.  All royalty payments required under this Paragraph shall be due within thirty (30) days of the date PHS provides Licensee notice of the payment due.

 

8.2                                Licensee agrees to have an audit of sales and royalties conducted by an independent auditor at least every two (2) years if annual sales of the Licensed Products or Licensed Processes or New Products are over two (2) million dollars.  The audit shall address, at a minimum, the amount of gross sales by or on behalf of Licensee during the audit period, terms of the license as to percentage or fixed royalty to be remitted to the Government , the amount of royalties owed to the Government under this Agreement , and whether the royalties owed have been paid to the Government and is reflected in the records of the Licensee .  The audit shall also indicate the PHS license number, product, and the time period being audited.  A report certified by the auditor shall be submitted promptly by the auditor directly to PHS and Licensee on completion.

 

9.                                       REPORTS ON PROGRESS, BENCHMARKS, SALES, AND PAYMENTS

 

9.1                                Prior to signing this Agreement , Licensee has provided PHS with the Commercial Development Plan in Appendix E, under which Licensee intends to bring the subject matter of the Licensed Patent Rights or New Products to the point of Practical Application.  This Commercial Development Plan is hereby incorporated by reference into this Agreement.  Based on this plan, performance Benchmarks are determined as specified in Appendix D.

 

9.2                                Licensee shall provide written annual reports on its product development progress or efforts to commercialize under the Commercial Development Plan for the Licensed Fields of Use within sixty (60) days after December 31 of each calendar year.  These progress reports shall include, but not be limited to: progress on research and development, status of applications for regulatory approvals, manufacturing, marketing, importing, and sales during the preceding calendar year, as well as, plans for the present calendar year .  PHS also encourages these reports to include information on any of Licensee’s public service activities that relate to the Licensed Patent Rights or New Products.  If reported progress differs from that projected in the Commercial Development Plan and

 



 

Benchmarks , Licensee shall explain the reasons for such differences In any annual report, Licensee may propose amendments to the Commercial Development Plan , acceptance of which by PHS may not be denied or delayed unreasonably.  Licensee agrees to provide any additional information reasonably required by PHS to evaluate Licensee ’s performance under this Agreement.  Licensee may amend the Benchmarks at any time upon written approval by PHS.  PHS shall not unreasonably withhold approval of any request of Licensee to extend the time periods of this schedule if the request is supported by a reasonable showing by Licensee of diligence in its performance under the Commercial Development Plan and toward bringing the Licensed Products or New Products to the point of Practical Application .

 

9.3                                Licensee shall report to PHS the dates for achieving Benchmarks specified in Appendix D and the First Commercial Sale in each country in the Licensed Territory within thirty (30) days of such occurrences.

 

9.4                                Licensee shall submit to PHS , within sixty (60) days after each calendar half-year ending June 30 and December 31, a royalty report, as described in the example in Appendix F, setting forth for the preceding half-year period the amount of the Licensed Products or New Products sold or Licensed Processes practiced by or on behalf of Licensee in each country within the Licensed Territory , the Net Sales, and the amount of royalty accordingly due With each royalty report, Licensee shall submit payment of earned royalties due If no earned royalties are due to PHS for any reporting period, the written report shall so state The royalty report shall be certified as correct by an authorized officer of Licensee and shall include a detailed listing of all deductions made under Paragraph 2.10 to determine Net Sales made under Article 6 to determine royalties due.

 

9.5                                Licensee agrees to forward semi-annually to PHS a copy of reports received by Licensee from its sublicensees during the preceding half-year period as shall be pertinent to a royalty accounting to PHS by Licensee for activities under the sublicense.

 

9.6                                Royalties due under Article 6 shall be paid in U.S. dollars and payment options are listed in Appendix G.  For conversion of foreign currency to U.S. dollars, the conversion rate shall be the New York foreign exchange rate quoted in The Wall Street Journal on the day that the payment is due, and any loss of exchange, value, taxes, or other expenses incurred in the transfer or conversion to U.S. dollars shall be paid entirely by Licensee.  The royalty report required by Paragraph 9.4 shall be mailed to PHS at its address for Agreement Notices indicated on the Signature Page.

 



 

9.7                                Licensee shall be solely responsible for determining if any tax on royalty income is owed outside the United States and shall pay this tax and be responsible for all filings with appropriate agencies of foreign governments.

 

9.8                                Additional royalties may be assessed by PHS on any payment that is more than ninety (90) days overdue at the rate of one percent (1%) per month.  This one percent (1%) per month rate may be applied retroactively from the original due date until the date of receipt by PHS of the overdue payment and additional royalties.  The payment of any additional royalties shall not prevent PHS from exercising any other rights it may have as a consequence of the lateness of any payment.

 

9.9                                All plans and reports required by this Article 9 and marked “confidential” by Licensee shall, to the extent permitted by law, be treated by PHS as commercial and financial information obtained from a person and as privileged and confidential, and any proposed disclosure of these records by the PHS under the Freedom of Information Act (FOIA), 5 U.S.C. §552 shall be subject to the predisclosure notification requirements of 45 CFR §5.65(d).

 

10.                                PERFORMANCE

 

10.1                         Licensee shall use its reasonable commercial efforts to bring the Licensed Products or New Products and Licensed Processes to Practical Application.  “Reasonable commercial efforts” for the purposes of this provision shall include adherence to the Commercial Development Plan in Appendix E and performance of the Benchmarks in Appendix D The efforts of a sublicense shall be considered the efforts of Licensee .

 

10.2                         Upon the First Commercial Sale , until the expiration or termination of this Agreement , Licensee shall use its reasonable commercial efforts to make Licensed Products or New Products and Licensed Processes reasonably accessible to the United States public.

 

10.3                         Licensee agrees, after its First Commercial Sale , to make reasonable quantities of Licensed Products or New Products ormaterialsproduced through the use of Licensed Processes available on a compassionate use basis to patients, either through the patient’s physician(s) or the medical center treating the patient.

 

10.4                         Licensee agrees, after its First Commercial Sale and as part of its marketing and product promotion, to develop educational materials (e.g., brochures, website, etc.) directed to patients and physicians detailing the Licensed Products or New Products or medical aspects of the prophylactic and therapeutic uses of the Licensed Products or New Products .

 

10.5                         Licensee agrees to supply, to the Mailing Address for Agreement Notices indicated on the Signature Page, the Office of Technology Transfer, NIH with

 



 

inert samples of the Licensed Products or New Products or their packaging for educational and display purposes only.

 

11.                                INFRINGEMENT AND PATENT ENFORCEMENT

 

11.1                         PHS and Licensee agree to notify each other promptly of each infringement or possible infringement of the Licensed Patent Rights , as well as, any facts which may affect the validity, scope, or enforceability of the Licensed Patent Rights of which either Party becomes aware.

 

11.2                         In the event that a declaratory judgment action alleging invalidity of any of the Licensed Patent Rights shall be brought against PHS , PHS agrees to notify Licensee that an action alleging invalidity has been brought.  PHS does not represent that it shall commence legal action to defend against a declaratory action alleging invalidity.  Licensee shall take no action to compel the Government either to initiate or to join in any declaratory judgment action.  Should the Government be made a party to any suit by motion or any other action of Licensee , Licensee shall reimburse the Government for any costs, expenses, or fees, which the Government incurs as a result of the motion or other action.  Upon Licensee’s payment of all costs incurred by the Government as a result of Licensee’s joinder motion or other action, these actions by Licensee shall not be considered a default in the performance of any material obligation under this Agreement .

 

12.                                NEGATION OF WARRANTIES AND INDEMNIFICATION

 

12.1                         PHS offers no warranties other than those specified in Article 1.

 

12.2                         PHS does not warrant the validity of the Licensed Patent Rights and makes no representations whatsoever with regard to the scope of the Licensed Patent Rights , or that the Licensed Patent Rights and Supplied Materials may be exploited without infringing other patents or other intellectual property rights of third parties.

 

12.3                         PHS MAKES NO WARRANTIES, EXPRESSED OR IMPLIED, OF MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE OF ANY SUBJECT MATTER DEFINED BY THE CLAIMS OF THE LICENSED PATENT RIGHTS OR TANGIBLE MATERIALS RELATED THERETO, INCLUDING BUT NOT LIMITED TO SUPPLIED MATERIALS .

 

12.4                         PHS does not represent that it shall commence legal actions against third parties infringing the Licensed Patent Rights or Supplied Materials .

 

12.5                         Licensee shall indemnify and hold PHS , its employees, students, fellows, agents, and consultants harmless from and against all liability, demands, damages,

 



 

expenses, and losses, including but not limited to death, personal injury, illness, or property damage in connection with or arising out of:

 

(a)                                  the use by or on behalf of Licensee , its sublicensees, its directors, employees, or third parties of any Licensed Patent Rights or Supplied Materials ; or

 

(b)                                  the design, manufacture, distribution, or use of any Licensed Products , Licensed Processes or New Products by Licensee , or other products or processes developed in connection with or arising out of the Licensed Patent Rights.  Licensee agrees to maintain a liability insurance program consistent with sound business practice.

 

12.6                         Licensee agrees to maintain a liability insurance program consistent with sound business practice.

 

13.                                TERM, TERMINATION, AND MODIFICATION OF RIGHTS

 

13.1                         This Agreement is effective when signed by all parties, unless the provisions of Paragraph 14.15 are not fulfilled, and shall extend to the expiration of the last to expire of the Licensed Patent Rights unless sooner terminated as provided in this Article 13.

 

13.2                         In the event that Licensee is in default in the performance of any material obligations under this Agreement , including but not limited to the obligations listed in Paragraph 13.05, and if the default has not been remedied within ninety (90) days after the date of notice in writing of the default, PHS may terminate this Agreement by written notice and pursue outstanding royalties owed through procedures provided by the Federal Debt Collection Act.

 

13.3                         In the event that Licensee becomes insolvent, files a petition in bankruptcy, has such a petition filed against it, determines to file a petition in bankruptcy, or receives notice of a third party’s intention to file an involuntary petition in bankruptcy, Licensee shall immediately notify PHS in writing Furthermore, PHS shall have the right to terminate this Agreement immediately upon Licensee’s receipt of written notice.

 

13.4                         Licensee shall have a unilateral right to terminate this Agreement in any country or territory by giving PHS sixty (60) days written notice to that effect.

 

13.5                         PHS shall specifically have the right to terminate or modify, at its option, this Agreement , if PHS determines that the Licensee :

 

(a)                                  is not executing the Commercial Development Plan submitted with its request for a license and the Licensee cannot otherwise demonstrate to PHS’ satisfaction that the Licensee has taken, or can be expected to take

 



 

within a reasonable time, effective steps to achieve Practical Application of the Licensed Products or New Products ;

 

(b)                                  has not achieved the Benchmarks as may be modified under Paragraph 9.2;

 

(c)                                   has willfully made a false statement of, or willfully omitted, a material fact in the license application or in any report required by this Agreement ;

 

(d)                                  has committed a material breach of a covenant or agreement contained in this Agreement ;

 

(e)                                   is not keeping Licensed Products or New Products reasonably available to the public after commercial use commences;

 

(f)                                    cannot reasonably satisfy unmet health and safety needs; or

 

(g)                                   cannot reasonably justify a failure to comply with the domestic production requirement of Paragraph 5.2, unless waived.

 

13.6                         In making the determination referenced in Paragraph 13.5, PHS shall take into account the normal course of such commercial development programs conducted with sound and reasonable business practices and judgment and the annual reports submitted by Licensee under Paragraph 9.2 Prior to invoking termination or modification of this Agreement under Paragraph 13.5, PHS shall give written notice to Licensee providing Licensee specific notice of, and a ninety (90) day opportunity to respond to, PHS’ concerns as to the items referenced in 13.5(a)-13.5(g) If Licensee fails to alleviate PHS’ concerns as to the items referenced in 13.5(a)-13.5(g) or fails to initiate corrective action to PHS ’ satisfaction, PHS may terminate this Agreement .

 

13.7                         PHS reserves the right according to 35 U.S.C. §209(d)(3) to terminate or modify this Agreement if it is determined that the action is necessary to meet the requirements for public use specified by federal regulations issued after the date of the license and these requirements are not reasonably satisfied by Licensee .

 

13.8                         Within thirty (30) days of receipt of written notice of PHS’ unilateral decision to modify or terminate this Agreement , Licensee may, consistent with the provisions of 37 CFR §404.11, appeal the decision by written submission to the designated PHS official The decision of the designated PHS official shall be the final agency decision.  Licensee may thereafter exercise any and all administrative or judicial remedies that may be available.

 

13.9                         Within ninety (90) days of expiration or termination of this Agreement under this Article 13, a final report shall be submitted by Licensee.  Any royalty payments, including those incurred but not yet paid (such as the full minimum annual royalty), and those related to patent expense, due to PHS shall become

 



 

immediately due and payable upon termination or expiration If terminated under this Article 13, sublicensees may elect to convert their sublicenses to direct licenses with PHS pursuant to Paragraph 4.3.  Unless otherwise specifically provided for under this Agreement , upon termination or expiration of this Agreement, Licensee shall return all Licensed Products and New Products or other materials included within the Licensed Patent Rights and under its control to PHS or provide PHS with written certification of the destruction thereof.

 

14.                                GENERAL PROVISIONS

 

14.1                         Neither party may waive or release any of its rights or interests in this Agreement except in writing.  The failure of the Government to assert a right hereunder or to insist upon compliance with any term or condition of this Agreement shall not constitute a waiver of that right by the Government or excuse a similar subsequent failure to perform any of these terms or conditions by Licensee .

 

14.2                         This Agreement constitutes the entire Agreement between the Parties relating to the subject matter of the Licensed Patent Rights , Licensed Products , New Products , Supplied Materials and Licensed Processes , and all prior negotiations, representations, agreements, and understandings are merged into, extinguished by, and completely expressed by this Agreement .

 

14.3                         The provisions of this Agreement are severable, and in the event that any provision of this Agreement shall be determined to be invalid or unenforceable under any controlling body of law, this determination shall not in any way affect the validity or enforceability of the remaining provisions of this Agreement .

 

14.4                         If either party desires a modification to this Agreement , the parties shall, upon reasonable notice of the proposed modification by the party desiring the change, confer in good faith to determine the desirability of the modification No modification shall be effective until a written amendment is signed by the signatories to this Agreement or their designees.

 

14.5                         The construction, validity, performance, and effect of this Agreement shall be governed by Federal law as applied by the Federal courts in the District of Columbia.

 

14.6                         All Agreement notices required or permitted by this Agreement shall be given by prepaid, first class, registered or certified mail or by an express/overnight delivery service provided by a commercial carrier, properly addressed to the other party at the address designated on the Signature Page, or to any other address as may be designated in writing by such other party.  Agreement notices shall be considered timely if such notices are received on or before the established deadline date or sent on or before the deadline date as verifiable by U.S. Postal Service postmark or dated receipt from a commercial carrier.  Parties should

 



 

request a legibly dated U.S. Postal Service postmark or obtain a dated receipt from a commercial carrier or the U.S. Postal Service.  Private metered postmarks shall not be acceptable as proof of timely mailing.

 

14.7                         This Agreement shall not be assigned by Licensee except:

 

(a)                                  with the prior written consent of PHS , this consent shall not to be withheld unreasonably; or

 

(b)                                  as part of a sale or transfer of substantially the entire business of Licensee relating to operations which concern this Agreement ; and

 

(c)                                   Licensee shall notify PHS within ten (10) days of any assignment of this Agreement by Licensee , and Licensee shall pay PHS , as an additional royalty, one (1) percent of the fair market value of any consideration received for any assignment of this Agreement within thirty (30) days of the assignment.

 

14.8                         Licensee agrees in its use of any Supplied Materials to comply with all applicable statutes, regulations, and guidelines, including PHS and HHS regulations and guidelines .  Licensee agrees not to use the Supplied Materials for research involving human subjects or clinical trials in the United States without complying with 21 CFR Part 50 and 45 CFR Part 46.  Licensee agrees not to use the Supplied Materials for research involving human subjects or clinical trials outside of the United States without notifying PHS , in writing, of the research or trials and complying with the applicable regulations of the appropriate national control authorities.  Written notification to PHS of research involving human subjects or clinical trials outside of the United States shall be given no later than sixty (60) days prior to commencement of the research or trials.

 

14.9                         Licensee acknowledges that it is subject to and agrees to abide by the United States laws and regulations (including the Export Administration Act of 1979 and Arms Export Control Act) controlling the export of technical data, computer software, laboratory prototypes, biological materials, Supplied Materials and other commodities.  The transfer of these items may require a license from the appropriate agency of the Government or written assurances by Licensee that it shall not export these items to certain foreign countries without prior approval of the agency.  PHS neither represents that a license is or is not required or that, if required, it shall be issued.

 

14.10                  Licensee agrees to mark the Licensed Products or New Products or their packaging sold in the United States with all applicable U.S. patent numbers and similarly to indicate “Patent Pending” status.  All Licensed Products or New

 



 

Products manufactured in, shipped to, or sold in other countries shall be marked in a manner to preserve PHS patent rights in those countries.

 

14.11                  By entering into this Agreement , PHS does not directly or indirectly endorse any product or service provided, or to be provided, by Licensee whether directly or indirectly related to this Agreement.  Licensee shall not state or imply that this Agreement is an endorsement by the Government , PHS , any other Government organizational unit, or any Government employee Additionally, Licensee shall not use the names of NIH, PHS, FDA or HHS or the Government or their employees in any advertising, promotional, or sales literature without the prior written approval of PHS .

 

14.12                  The Parties agree to attempt to settle amicably any controversy or claim arising under this Agreement or a breach of this Agreement , except for appeals of modifications or termination decisions provided for in Article 13 .  Licensee agrees first to appeal any unsettled claims or controversies to the designated PHS official, or designee, whose decision shall be considered the final agency decision.  Thereafter, Licensee may exercise any administrative or judicial remedies that may be available.

 

14.13                  Nothing relating to the grant of a license, nor the grant itself, shall be construed to confer upon any person any immunity from or defenses under the antitrust laws or from a charge of patent misuse, and the acquisition and use of rights pursuant to 37 CFR Part 404 shall not be immunized from the operation of state or Federal law by reason of the source of the grant.

 

14.14                  Paragraphs 8.1, 9.7-9.9, 12.1-12.5, 13.8, 13.9, 14.12 and 14.14 of this Agreement shall survive termination of this Agreement .

 

14.15                  The terms and conditions of this Agreement shall, at PHS’ sole option, be considered by PHS to be withdrawn from Licensee’s consideration and the terms and conditions of this Agreement , and the Agreement itself to be null and void, unless this Agreement is executed by the Licensee and a fully executed original is received by PHS within sixty (60) days from the date of PHS signature found at the Signature Page.

 

SIGNATURES BEGIN ON NEXT PAGE

 



 

PHS PATENT LICENSE AGREEMENT — NONEXCLUSIVE

 

SIGNATURE PAGE

 

For PHS:

 

 

 

 

 

/s/ Steven M. Ferguson

 

4/25/07

Steven M. Ferguson

 

Date

Director, Division of Technology Development and Transfer

 

 

Office of Technology Transfer

 

 

National Institutes of Health

 

 

 

Mailing Address for Agreement notices:

 

Chief, Monitoring & Enforcement Branch
Office of Technology Transfer
National Institutes of Health
6011 Executive Boulevard, Suite 325
Rockville, Maryland 20852-3804 U.S.A.

 

For Licensee (Upon, information and belief, the undersigned expressly certifies or affirms that the contents of any statements of Licensee made or referred to in this document are truthful and accurate.):

 

by:

 

 

 

 

 

/s/ Ronald H.W. Lorijn

 

5/2/07

Signature of Authorized Official

 

Date

 

 

 

Ronald H.W. Lorijn

 

 

Printed Name

 

 

 

 

 

C.E.O.

 

 

Title

 

 

 

I.                                         Official and Mailing Address for Agreement notices:

Sander van Deventer, M.D.

Chief Scientific Officer

Amsterdam Molecular Therapeutics

Meibergdreef 61

P.O. Box 22506

1100DA Amsterdam, Netherlands

 



 

II.                                    Official and Mailing Address for Financial notices ( Licensee’s contact person for royalty payments)

 

Sander van Deventer, M.D.

Name

 

Chief Scientific Officer

Title

 

Mailing Address:

 

Amsterdam Molecular Therapeutics

Meibergdreef 61

P.O. Box 22506

1100DA Amsterdam, Netherlands

 

Email Address:                                        s.vandeventer@amtbv.com

Phone:                                                                                    +31-20-5669272

Fax:                                                                                                 +31-20-5669272

 

Any false or misleading statements made, presented, or submitted to the Government , including any relevant omissions, under this Agreement and during the course of negotiation of this Agreement are subject to all applicable civil and criminal statutes including Federal statutes 31 U.S.C. §§3801-3812 (civil liability) and 18 U.S.C. §1001 (criminal liability including fine(s) and/or imprisonment).

 



 

APPENDIX A - PATENT(S) OR PATENT APPLICATION(S)

 

Patent(s) or Patent Application(s):

 

I.                                         U.S. Patent Application(s) or Patent(s):

 

(a)  U.S. Patent Application Serial No. 09/986,618, filed November 9, 2001, now abandoned, entitled “Production of Adeno-Associated Virus in Insect Cells” [HHS Ref. No. E-325-2001/0-US-01];

(b)  U.S. Patent No. 6,723,551, issued April 20, 2004, entitled “Production of Adeno-Associated Virus in Insect Cells” [HHS Ref. No. E-325-2001/1-US-01]; and

(c)  U.S. Patent Application Serial No. 10/415,834, filed May 2, 2003, entitled “Production of Adeno-Associated Virus in Insect Cells” [HHS Ref. No. E-325-2001/2-US-02].

 

II.                                    PCT or Foreign Patent Application(s) or Patent(s):

 

(a)  PCT Patent Application Serial No. PCT/US02/35829, filed November 8, 2002, now abandoned, entitled “Production of Adeno-Associated Virus in Insect Cells” [HHS Ref. No. E-325-2001/2-PCT-01];

(b)  Canadian Patent Application Serial No. 2,467,959, filed May 6, 2004, entitled “Production of Adeno-Associated Virus in Insect Cells” [HHS Ref. No. E-325-2001/2-CA-03];

(c)  European Patent Application Serial No. 02795604.4, filed June 8, 2004, entitled “Production of Adeno-Associated Virus in Insect Cells” [HHS Ref. No. E-325-2001/2-EP-04]; and

(d)  Australian Patent Application Serial No. 2002360355, filed April 28, 2004, entitled “Production of Adeno-Associated Virus in Insect Cells” [FIHS Ref. No. E-325-2001/2-AU-05].

 



 

APPENDIX B - LICENSED FIELDS OF USE AND TERRITORY

 

I.                                         Licensed Fields of Use :

 

Use of the Licensed Patent Rights for the commercial development of AAV related products within the scope of the Agreement .

 

II.                                    Licensed Territory :

 

United States, Australia, Canada and Europe.

 



 

APPENDIX C - ROYALTIES

 

Royalties:

 

I.                                         Licensee agrees to pay to PHS a nonereditable, nonrefundable license issue royalty in the amount of twelve thousand U.S. dollars ($12,000) within thirty (30) days from the effective date of this Agreement .

 

II.                                    Licensee agrees to pay to PHS a nonrefundable minimum annual royalty in the amount of fifteen thousand U.S. dollars ($15,000) as follows:

 

(a)                                  The first minimum annual royalty is due within thirty (30) days of the effective date of this Agreement and may be prorated according to the fraction of the calendar year remaining between the effective date of this Agreement and the next subsequent January 1; and

 

(b)                                  Subsequent minimum annual royalty payments are due and payable on January 1 of each calendar year and may be credited against any earned royalties due for sales made in that year.

 

III.                               Licensee agrees to pay PHS earned royalties of one percent (1%) on Net Sales by or on behalf of Licensee for Licensed Products .

 

IV.                                Licensee agrees to pay PHS earned royalties of one quarter of one percent (.25%) on Net Sales by or on behalf of Licensee for New Products .

 

V.                                     Licensee agrees to pay PHS Benchmark royalties within thirty (30) days of achieving each Benchmark :

 

(a)                                  Thirty thousand U.S. dollars ($30,000) — Initiation of each Phase I clinical trial or foreign equivalent.

 

(b)                                  Seventy-five thousand U.S. dollars ($75,000) — Initiation of each Phase II clinical trial or foreign equivalent.

 

(c)                                   One hundred and fifty thousand U.S. dollars ($150,000) — Initiation of each Phase III clinical trial or foreign equivalent.

 

(d)                                  Initiation of first Marketing Approval or foreign equivalent in the following jurisdictions/countries:

 

(i)                                            Three hundred thousand U.S. dollars ($300.000) in Europe.

 

(ii)                                         Three hundred thousand U.S. dollars ($300,000) in the United States.

 

(iii)                                      Seventy-five thousand U.S. dollars ($75,000) in Australia.

 

(iv)                                     Seventy-five thousand U.S. dollars ($75,000) in Canada.

 



 

IV.                                Licensee agrees to pay PHS additional sublicensing royalties as follows:

 

Ten percent (10%) of the fair market value of any consideration received for granting each sublicense.

 



 

APPENDIX D - BENCHMARKS AND PERFORMANCE

 

Licensee agrees to the following Benchmarks for its performance under this Agreement and, within thirty (30) days of achieving a Benchmark , shall notify PHS that the Benchmark has been achieved.

 

I.                                         Initiation of Preclinical Development phase or foreign equivalent — 4 th  quarter 2006.

 

II.                                    Initiation of first Phase I clinical trial or foreign equivalent — 3 rd quarter 2007.

 

III.                               Initiation of first Phase II clinical trial or foreign equivalent — 3 rd  quarter 2007.

 

IV.                                Initiation of first Phase III clinical trial or foreign equivalent — 4 th  quarter 2007.

 

V.                                     Submission to Regulatory Authority of first Marketing Approval or foreign equivalent — 2 nd  quarter 2008.

 



 

APPENDIX E - COMMERCIAL DEVELOPMENT PLAN

 

Introduction

 

In 2004 the European Regulatory Authority (“EMEA”), assigned the ‘orphan drug’ status to Licensee’s lead product AMT-010.  AMT-010 (Adeno-Associated Viral Vector Expressing Human Lipoprotein LipaseS447X), a gene therapy product to treat Lipoprotein Lipase Deficiency Type I and V deficiency, has entered its phase I/II clinical stage.  This program is focused on treating the rare, but serious and disabling inherited disease called LPL type 1 deficiency for which no adequate treatment exists today.

 

Due to a substantial change in the manufacturing process (from DKFZ to Baculo), Licensee does not intend to market AMT-010 but instead the newly produced product AMT-011.

 

For that reason, Licensee has started preclinical development in 2006, and will initiate a phase II clinical study with AMT-011 in Canada in Q3 2007, as well as a phase II clinical study for type V hyperlipoproteinemia in Q4 2007 .  Licensee expects to file the registration dossier for type I hyperlipoproteinemia with the EMEA by Q1 2008.

 

Early 2007, Licensee will begin discussions with the FDA to prepare the filing of the AMT-011 dossier for registration in the U.S.A.  Depending on the feedback from the U.S. authorities, Licensee plans to file its registration dossier with the FDA in 2008.

 

Technology

 

Licensee is building gene therapies using adeno-associated viral (AAV)-based vectors These vectors do not integrate into the host genome and result in long-lasting expression of therapeutic genes.  AAV vectors can be specifically targeted to various organs (i.e. muscle, brain, liver, retina) and even to specific cells within a target organ.  Improvements in local expression are also achieved by using potent organ specific promoters.  Licensee has extensively optimized expression of therapeutic genes in various organs by selection of AAV serotype-promoter combinations and by selection of high expressing transgenes.  In relevant preclinical models, this has resulted in lifelong (2 years for mouse and rat) expression of transgenes at therapeutic levels (often 100% of the normal expression) and a complete cure of the disease.  Because of this extensive knowledge base, and the availability of all relevant currently available AAV serotypes, Licensee believes it is in the position to rapidly develop genetic therapies for a wide range of diseases caused by single gene defects.

 

The Disease: Lipoprotein Lipase Deficiency

 

Genetic lipoprotein lipase (LPL) deficiency results in profound hypertriglyceridemia, which is associated with intense chronic abdominal pain, hepatosplenomegaly, eruptive xanthomas, lipemia retinalis, dyspnea, mono- or polyparesthesias, and memory loss.  Prolonged elevations in plasma triglycerides (TG) also induce recurrent episodes of often lethal pancreatitis, chronic pancreatic insufficiency, and diabetes mellitus.  Currently, no effective treatment for this disease exists.  Patients must follow a strict low-fat diet.  However, TG levels often remain above the

 



 

critical threshold.  Genetic LPL deficiency type I is a rare, autosomal recessive trait.  Prevalence varies between 1 in 1,000,000 in the general population to 1 in 5,000 in French Quebec (a ‘founder effect’).

 

LPL gene therapy will also be investigated to treat type V hyperlipoproteinemia (prevalence of 1.8 in 10,000).  LPL gene therapy may improve the quality of life and reduce the risk of morbidity and mortality for a significant number of patients that suffer from this particular lipid disorder.

 

Currently, no treatment for hyperlipoproteinemia is available, and patients suffer from repeated bouts of pancreatitis.  LPL enzyme replacement therapy is not feasible in view of the very short half-life of the enzyme.  The only advice a physician can give these patients is to keep a strict fat-free diet, which is extremely difficult to maintain.  Further, even with such a diet, the serum triglyceride levels remain far above the critical level of 10 µmol/L.

 

Business Strategy

 

Licensee’s core strategy will be to position the new medicine as an orphan drug This strategy has several important benefits.  The Regulatory Authorities in many countries including the EU (EMEA) and the U.S.A. (FDA) have recognized the significance of the development of orphan drugs.  Thanks to the regulatory laws and regulations the ‘time-to-market’, IP and marketing rights protection are very favorable for such products.

 

Marketing exclusivity for orphan drugs after marketing authorization are:

 

·                   EU: 10 years

·                   USA: 7 years

 

This means that during that period no other sponsor can obtain marketing authorization for a similar product in the designated indication.  Licensee received the orphan drug status for the LPL gene therapy from the EMEA last year.  Due to these circumstances and the fact that the major pharmaceutical companies have little or no interest in developing products for these niche markets, the opportunities for companies such as Licensee are significant.

 

In the specific case for the LPL product, there is yet another element that will ensure a quick uptake and fast penetration in the target market.  Specifically, there is no treatment today nor in the foreseeable future for patients suffering from LPL Type 1 and V deficiency.  In other words, no major competing treatments are available to these patients.

 

In conclusion, no cure or symptomatic treatment, which would alleviate the disease’s symptoms and its complications exist today.  The LPL project is unique because it exploits for the first time the possibility to treat patients suffering from LPL deficiency type I and V.

 

Marketing and Sales

 

Market Overview AMT-011

 

Lipoprotein lipase deficiency Type I

 

Hypertriglyceridemia Type V

 



 

·              Orphan indication

 

·              Larger indication

·              3,000-4,000 patients world-wide; very well organized and active patients groups

 

·              30% of the hypertriglyceridemia patients have underlying LPL deficiency

·              Estimated global sales $ 200 M

 

·              Estimated global sales >$500 M

·              No competition

 

·              Competition: small molecule approaches

 

Licensee’s business strategy focuses on market entry for its orphan indications through its own dedicated marketing and sales force in Europe and North America.  In the Western world, many patients suffering from serious orphan diseases have formed patient groups.  Also their treating physicians, in many instances, are well connected.  After marketing approval has been obtained, this situation allows for a very concentrated educational effort to inform patients and physicians about the important benefits of gene therapy.  In order to ethically justify and successfully penetrate such markets, Licensee will ensure to build-up a highly educated medical service team to assist physicians in the selection of those patients who will benefit from the treatment.  Such a team can be relatively small, which allows management to monitor and guide it closely.

 



 

APPENDIX F - EXAMPLE ROYALTY REPORT

 

Required royalty report information includes :

 

·                   OTT license reference number (L-XXX-200X/0)

·                   Reporting period

·                   Catalog number and units sold of each Licensed Product (domestic and foreign)

·                   Gross Sales per catalog number per country

·                   Total Gross Sales

·                   Itemized deductions from Gross Sales

·                   Total Net Sales

·                   Earned Royalty Rate and associated calculations

·                   Gross Earned Royalty

·                   Adjustments for Minimum Annual Royalty (MAR) and other creditable payments made

·                   Net Earned Royalty due

 

Example

 

Catalog Number

 

Product
Name

 

Country

 

Units Sold

 

Gross Sales
(US$)

 

1

 

A

 

US

 

250

 

62,500

 

1

 

A

 

UK

 

32

 

16,500

 

1

 

A

 

France

 

25

 

15,625

 

2

 

B

 

US

 

0

 

0

 

3

 

C

 

US

 

57

 

57,125

 

4

 

D

 

US

 

12

 

1,500

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Total Gross Sales

 

153,250

 

 

 

 

 

 

 

Less Deductions:

 

 

 

 

 

 

 

 

 

Freight

 

3,000

 

 

 

 

 

 

 

Returns

 

7,000

 

 

 

 

 

 

 

Total Net Sales

 

143,250

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Royalty Rate

 

8

%

 

 

 

 

 

 

Royalty Due

 

11,460

 

 

 

 

 

 

 

Less Creditable Payments

 

10,000

 

 

 

 

 

 

 

Net Royalty Due

 

1,460

 

 



 

APPENDIX G - ROYALTY PAYMENT OPTIONS

 

NIH/PHS License Agreements

 

*In order to process payment via Electronic Funds Transfer sender MUST supply the following information:

 

Procedure for Transfer of Electronic Funds to NIH for Royalty Payments

 

Bank Name:  Federal Reserve Bank

 

ABA# 021030004
TREAS NYC
BNF=/AC-75080031
OBI=Licensee Name and OTT Reference Number
Dollar Amount Wired=$$

 

NOTE: Only U.S. banks can wire directly to the Federal Reserve Bank.  Foreign banks cannot wire directly to the Federal Reserve Bank, but must go through an intermediary U.S. bank.  Foreign banks may send the wire transfer to the U.S. bank of their choice, who, in turn forwards the wire transfer to the Federal Reserve Bank.

 

Mailing Address for Royalty Payments:

 

National Institutes of Health
P.O. Box 360120
Pittsburgh, PA 15251-6120 USA

 

Overnight Mail for Royalty Payments only

 

National Institutes of Health
360120
Mellon Client Service Center
Room 670
500 Ross Street
Pittsburgh, PA 15262-0001

 

(412) 234-4381 (Customer Service)

 

Please make checks payable to: NIH/Patent Licensing

 

The OTT Reference Number MUST appear on checks, reports and correspondence

 



 

PUBLIC HEALTH SERVICE

 

FIRST AMENDMENT TO L-107-2007/0

 

This is the first amendment (“ First Amendment ”) of the agreement by and between the National Institutes of Health (“ NIH ”) or the Food and Drug Administration (“ FDA ”), hereinafter singly or collectively referred to as agencies of the United States Public Health Service (“ PHS ”) within the Department of Health and Human Services (“ HHS ”), and Amsterdam Molecular Therapeutics having an effective date of May 2, 2007 and having NIH Reference Number L-107-2007/0 (“ Agreement ”).  This First Amendment , having NIH Reference Number L-107-2007/1, is made between the PHS through the Office of Technology Transfer, NIH , having an address at 6011 Executive Boulevard, Suite 325, Rockville, Maryland 20852-3804, U.S.A., and Amsterdam Molecular Therapeutics, having an office at Meibergdreef 61, 1005 BA Amsterdam, The Netherlands (“ Licensee ”).  This First Amendment includes, in addition to the amendments made below, 1) a Signature Page, 2) Appendix D — Benchmarks and Performance , 3) Appendix E — Commercial Development Plan , 4) Exhibit A — Request to Collaborate with Institute Pasteur, 5) Exhibit B- PHS Consent for Institute Pasteur Exemption, and 6) Attachment 1 (Royalty Payment Information).

 

WHEREAS, Licensee has requested an amendment to revise the Benchmarks for its lead Licensed Product AMT-011, also known as Glybera™;

 

WHEREAS, Licensee did not conduct a Phase III clinical trial for Glybera™ and has filed for Marketing Approval for Glybera™ with the European Medicines Agency on January 11, 2010;

 

WHEREAS, Licensee has requested Benchmark royalty exemption for collaborations with not-for-profit organizations and academic institutions for pre-clinical and clinical development to treat ultra-orphan indications;

 

WHEREAS, PHS requested that Licensee amend the Appendix E - Commercial Development Plan to state its development plans for Licensed Products other than Glybera™;

 

WHEREAS, PHS requested that Licensee add Benchmarks for Licensed Products other than Glybera™ to Appendix D - Benchmarks and Performance ;

 

WHEREAS, PHS and Licensee desire that the Agreement be amended a first time as set forth below in order amend Appendix D - Benchmarks and Performance and Appendix E - Commercial Development Plan .

 

NOW, THEREFORE, in consideration of the mutual covenants and promises contained herein, PHS and Licensee , intending to be bound, hereby mutually agree to the following:

 

1)              The following modifications shall be made to the Agreement:

 

a.               The following Paragraphs 2.14, 2.15, 2.16, and 6.8 shall be added to the Agreement :

 

2.14                         Orphan Indication ” means a disease that affects less than two hundred thousand (200,000) people in the United States as defined by the Food and

 



 

Drug Administration or five (5) in ten thousand (10,000) people in the European Union as defined by the European Medicines Agency.

 

2.15                         Ultra-Orphan Indication ” means a disease that effects less than one (1) in Fifty Thousand (50,000) people in the United States or the European Union.

 

2.16                         Exempt Collaborator ” means a not-for-profit organization or academic institution that has entered a formal collaboration and / or supply agreement with Licensee to conduct pre-clinical development and solely sponsor clinical trials of Licensed Product , excluding Supplied Materials , to treat an Ultra-Orphan Indication ; in which Licensee may acquire clinical development and data for regulatory approval and sale of a Licensed Product .

 

6.8                                Unless otherwise exempted in Article 15, Licensee agrees to pay PHS benchmark royalties as set forth in Appendix C.

 

b.               Paragraph 6.3 shall be deleted from the Agreement and replaced with the following Paragraph 6.3:

 

6.3                                Unless otherwise exempted in Article 15, Licensee agrees to pay PHS earned royalties as set forth in Appendix C.

 

c.                Appendix D - Benchmarks and Performance of the Agreement shall be deleted and replaced with the Appendix D - Benchmarks and Performance attached to this First Amendment .

 

d.               Appendix E - Commercial Development Plan attached to this First Amendment shall be added the Agreement .

 

e.                The following Article 15 shall be added to the Agreement :

 

15.                                EXEMPTION FOR ULTRA-ORPHAN INDICATION RESEARCH

 

15.1                         Licensee shall be permitted, upon PHS consent, (not to be unreasonably withheld), to manufacture and supply Licensed Product , excluding Supplied Materials , to an Exempt Collaborator for use solely in pre-clinical and clinical development to treat an Ultra-Orphan Indication .  Prior to commencement of manufacturing of Licensed Product for an Exempt Collaborator , Licensee shall request permission in writing and must obtain written consent from PHS .  Additional documentation to establish an Exempt Collaborator may be required by PHS .

 

For avoidance of doubt, Licensee shall retain Supplied Materials and shall not release Supplied Materials to an Exempt Collaborator .

 

15.2                         Upon receipt of written consent from PHS for manufacturing of a Licensed Product for an Exempt Collaborator , Licensee shall not be

 



 

obligated to pay Benchmark royalties which would have been payable under Appendix C , Section V for Benchmarks triggered by clinical trials solely sponsored by the Exempt Collaborator until such time as Licensee exercises its option to acquire the clinical development from the Exempt Collaborator .

 

15.3                         Upon acquisition of the clinical development from an Exempt Collaborator , Licensee shall pay PHS royalties which become payable from that point onwards in accordance with Appendix C , Section V.  Licensee must inform PHS in writing within thirty (30) days of Licensee’s decision to acquire or not acquire clinical development from the Exempt Collaborator .

 

For avoidance of doubt, PHS shall consider Licensee’s sponsorship or co-sponsorship of a clinical trial or regulatory submission for a Licensed Product to treat an Ultra-Orphan Indication as an acquisition of clinical development from an Exempt Collaborator .

 

15.4                         Earned royalty payments on Net Sales specified in Appendix C, Section III shall not be applicable to Licensed Product manufactured for research and clinical trials conducted by an Exempt Collaborator approved by PHS per Paragraph 15.1.

 

In lieu of earned royalty payments, Licensee shall pay PHS a royalty payment of ten thousand dollars ($10,000) for each collaboration with an Exempt Collaborator approved by PHS .  Such royalty shall be due within thirty (30) days of the date of PHS written consent per Paragraph 15.1.

 

In case several PHS licenses apply to the same Licensed Product , only a single payment of $10,000 shall be payable per collaboration.

 

2)              Within sixty (60) days of the execution of this First Amendment , Licensee shall pay PHS an amendment issue royalty in the sum of One Thousand US Dollars ($1,000.00), to be sent to the address specified in Attachment 1.

 

3)              In the event any provision(s) of the Agreement is/are inconsistent with Attachment 1, such provision(s) is/are hereby amended to the extent required to avoid such inconsistency and to give effect to the payment information in such Attachment 1.

 

4)              All terms and conditions of the Agreement not herein amended remain binding and in effect.

 

5)              The terms and conditions of this Amendment shall, at PHS’ sole option, be considered by PHS to be withdrawn from Licensee’s consideration and the terms and conditions of this Amendment , and the Amendment itself to be null and void, unless this Amendment is executed by the Licensee and a fully executed original is received by PHS within sixty (60) days from the date of PHS signature found at the Signature Page.

 



 

6)              This First Amendment is effective on December 31, 2009 upon execution by all parties.

 

SIGNATURES BEGIN ON NEXT PAGE

 



 

The Netherlands

 

Email Address: p.morgan@amtbiopharma.com

 

 

Phone:

+31(0)20 566 7509

 

 

Fax:

+31(0)20 566 9272

 

 

II.    Official and Mailing Address for Financial notices (Licensee’s contact person for royalty payments):

 

 

Piers Morgan

Name

 

Chief Financial Officer

Title

 

Mailing Address:

 

Meibergdreef 61

 

1105 BA Amsterdam

 

The Netherlands

 

 

Email Address: p.morgan@ amtbiopharma.com

 

 

Phone:

+31(0)20 566 7509

 

 

Fax:

+31(0)20 566 9272

 

Any false or misleading statements made, presented, or submitted to the Government , including any relevant omissions, under this Agreement and during the course of negotiation of this Agreement are subject to all applicable civil and criminal statutes including Federal statutes 31 U.S.C. §§3801-3812 (civil liability) and 18 U.S.C. §1001 (criminal liability including fine(s) or imprisonment).

 



 

APPENDIX D - BENCHMARKS AND PERFORMANCE

 

Licensee agrees to the following Benchmarks for its performance under this Agreement and, within thirty (30) days of achieving a Benchmark, shall notify PHS that the Benchmark has been achieved.

 

Benchmarks for Lead Licensed Product (AMT-011 also known as Glybera™)

 

I.                                         Initiation of Preclinical Development phase or foreign equivalent - 4 th  quarter 2006.

 

II.                                    Initiation of first Phase I clinical trial or foreign equivalent - 3 rd  quarter 2007.

 

III.                               Initiation of first Phase II clinical trial or foreign equivalent - 3 rd  quarter 2007.

 

IV.                                Submission to Regulatory Authority of first Marketing Approval or foreign equivalent - l stl  quarter 2010.

 

Benchmarks for other Orphan Indication Licensed Product s (AMT-021 or equivalent)

 

I.                                         Initiation of Preclinical Development phase or foreign equivalent - Q4 2010

 

II.                                    Initiation of first Phase I clinical trial or foreign equivalent - Q3 2012

 

III.                               Initiation of first Phase II clinical trial or foreign equivalent - Q3 2014

 

IV.                                Submission to Regulatory Authority of first Marketing Approval or foreign equivalent - Q3 2015

 

Benchmarks for Licensed Products (AMT-090 or equivalent)

 

I.                                         Initiation of Preclinical Development phase or foreign equivalent - Q3 2011

 

II.                                    Initiation of first Phase I clinical trial or foreign equivalent - Q3 2012

 

III.                               Initiation of first Phase II clinical trial or foreign equivalent - Q3 2014

 

IV.                                Initiation of first Phase III clinical trial or foreign equivalent- Q3 2017

 

V.                                     Submission to Regulatory Authority of first Marketing Approval or foreign equivalent - Q3 2020

 



 

APPENDIX E - COMMERCIAL DEVELOPMENT PLAN

 

AMT-021 for Acute Intermittent Porphyria

 

Acute intermittent porphyria (AlP) is an autosomal dominant inherited condition caused by mutations in the porphobilinogen deaminase (PBGD) gene.  The PBGD gene is located on chromosome 11 q24.1 -24.2 and spread over fifteen exons.  The protein encoded by this gene is a rate-limiting enzyme, the PBGD enzyme, in the haem synthetic pathway.

 

More than 225 mutations of the PBGD gene have been described, all of them associated with loss of catalytic function.  The disease shows incomplete penetrance and only 20-50% of persons with one or more of the described mutations exhibit clinical symptoms of the disease.  The genetic disorder results in a 50% reduction of PBGD enzymatic activity.  This reduction of hepatic PBGD activity leads to an accumulation of toxic metabolites resulting from the blockade within the haem synthesis pathway.  Concentrations of haem precursors porphobilinogen (PGB) and delta-aminolevulinic acid (ALA) increase in blood and urine.  Lack of haem and/or accumulation of these metabolites are responsible for the acute attacks characteristic of this disease (Kauppinen et al 2005; Herrick and McColl 2005).  Currently, there is no treatment available for the disease.

 

Over the last couple of years we have explored AMT-021 (replication defective recombinant adeno-associated viral vector, AAV, containing the porphobilinogen deaminase gene) for therapeutic intervention in AlP.  AMT-021 is an AAV with pseudotype 5 capsid, which expresses the human PBGD gene under the transcriptional control of a liver specific promoter.  The therapeutic expression cassette consists of the human PBGD cDNA (codon optimised for human expression) inserted downstream of the liver specific promoter EalbAAT and upstream of a human PBGD polyadenylation sequence.

 

AMT-021 acts by delivering the PBGD expression cassette directly into hepatocytes.  The increase of PBGD enzymatic activity in the liver of AlP patients will provide sufficient enzyme to prevent the accumulation of toxic metabolites and thus, prevent porphyric attacks.

 

The aim of the project is to bring AAV5-PBDG therapy to patients.  AMT has already secured orphan designation for AAV5-PBDG treatment for AlP in Europe.  The table below describes the outline development plans, starting from a research batch production, and moving through to primate proof-of-concept, tox batch, pre-observational study, product development, GMP production, Phase I/II clinical trial, Phase lI/IIl clinical trial, all the way to regulatory filing.  Please note that the timelines are preliminary only, and that it is the nature of scientific and clinical development that planned timelines may change.

 

Preliminary Project Plan (Acute Intermittent Porphyria):

 

Task

 

Timelines

Research batch

 

2010

PoC in non-human primate

 

2010

Tox batch

 

2010

Toxicology (12 months)

 

2011

Pre-observation study

 

2011-2012

 



 

Clinical batch

 

2011

Interventional Phase I/II clinical trial

 

2012

2nd Observational study & Phase II/III clinical trial

 

2014

File with EMA

 

2015

Market Launch

 

2017

 

Project Plan Details:

 

The aim of this project is to develop a gene therapy product for the treatment of AlP, and to deliver a data package that is suitable for the submission and approval by the European and North American regulatory authorities.

 

Vector development and manufacturing

 

To develop a gene therapy for PBGD deficient patients, AAV5-PBDG product was designed to expresses the human PBGD gene under the control of a liver specific promoter.  AAV5-PBDG was produced in insect cells using the recombinant baculovirus method; sufficient amount of material was produced for efficacy studies in mice.  Methods to determine the quantity and purity of the rAAV batches were developed.  A purification process including chromatography and filtration steps was developed, further optimization and characterization of the scale-up procedure will be performed before a final batch for toxicology, for proof of principle and for clinical trials can be produced.

 

PoC in pre-clinical models

 

Because total deficiency of PBGD is lethal in mice, a compound heterozygous mouse (PBGD +/-  referred to as AlP mice) with ~35% of normal hepatic PBGD activity, has been developed as an established model to study AlP.  This murine model of AlP exhibits, after disease induction with phenobarbital (Pb), the typical biochemical characteristics of human AlP, notably, decreased hepatic PBGD activity, massively increased urinary excretion of haem precursors (ALA and PBG) and decreased motor function.

 

AlP mice were used to test the AAV5-PBDG product.  The therapeutic effect was evaluated three month after a single intravenous administration of AAV5-PBDG.  Efficacy of the therapy was demonstrated as the treatment was able to prevent disease induction with Pb. ALA and PBG levels in treated animals was reduced, and motor disturbance induced by Pb treatment, as measured in the Rotarod test, was almost completely abolished.  In addition, PBGD enzymatic activity increased in the AAV5-PBDG treated group 10 times over that of the control group.

 

This initial PoC will be repeated with the final version of the therapeutic vector following the completion of the vector development and manufacturing optimisation.  The final PoC will include the following:

 

·                   PoC in rodent disease model

·                   PoC in non-human primates, based on agreed protocol

 



 

GLP Toxicology

 

The aim of this section is to deliver toxicology study report suitable for the submission the regulatory authority.  The work will entail the following:

 

·                   Scientific advice from a regulatory body (AEMPS and/or EMA) for safety and toxicology package

·                   GLP toxicology study in rodents rats or mice, including any required biodistribution studies

·                   Supportive data for toxicology study in non-human primates

·                   GLP germline transmission study

 

Toxicology study design will take into account:

 

·                   Identification of potential target organs of biological activity and of potential target organs of toxicity

·                   Eventual concomitant medication (e.g. immunosuppressants, standard co-medication)

·                   Environmental risk/shedding

·                   Analysis of appropriateness of surrogate markers of efficacy/safety

·                   Any other relevant issues as may be identified

 

Clinical observational, pre-intervention study/studies


Before entering the interventional clinical study, an observation clinical study will be conducted to provide baseline information on the course of the disease by recording episodes AlP, abdominal pain, hospitalizations, extent of any possible known or unknown to be related to AlP symptomatology, incidence of (adverse) clinical events per year, etc. Sufficient data will be collected to provide a clinical picture to obtain a baseline data and to determine how efficacy will be shown during the interventional clinical trial.

 

Phase I/II


The clinical phase I/II should include an estimated minimum 6 patients that are administered the gene therapy drug, and are followed up and clinically assessed for at least 6 months following drug administration.  The primary aim of the clinical study will be safety and efficacy of the AAV5-PBDG product.  The clinical trial will include all biochemical, imaging, clinical and functional assays to assess the disease state and change therein over time, the phenotypic disease variation, as well as the overall clinical and psychosocial or other health status or change therein over time of the individual trial subjects, both before, during and following drug administration.

 

Phase II/III & Regulatory submission


After successful completion of Phase I/II study a Phase lI/IIl trial will be conducted with the aim of bringing the AlP therapy to market.  We estimate that 20-30 patients in total would be sufficient for regulatory filing of this product, as AlP is an ultra-orphan disease with a very limited patient number world-wide.

 

AMT-090 for Parkinson’s Disease

 

Parkinson’s disease (PD) is a progressive neurodegenerative disease, resulting in tremors, stiffness, slowness of movement, and lack of coordination.  Patients are faced with a severely debilitating disease and a serious loss in quality of life.  PD is caused by degeneration and death

 



 

of nerve cells in a specific part of the brain known as the substantia nigra.  These cells produce dopamine, a substance necessary for communication between nerve cells involved in the coordination of movement.

 

PD is the second most common neurodegenerative disease.  It usually affects people over 65, with an estimated total of 4.5 million patients worldwide.  Due to increasing life expectancy of the general population, the number of patients with PD is expected to double to around 9 million patients between now and 2030.

 

An ideal therapy for PD would decrease disability and slow down or halt disease progression.  Unfortunately, such treatments are not available yet and current therapies are limited to symptomatic treatment only.  These include levodopa, dopamine agonists, monoamine oxidase B (MAO-B) inhibitors and anticholinergic agents.

 

Glial cell line-derived neurotrophic factor (GDNF) was shown to promote the survival and differentiation of dopaminergic neurons.  The therapy aims to protect and enhance the function of the dopamine-producing nerve cells in the brain.  To date a number of clinical trials have been conducted in which recombinant GDNF protein has been directly delivered to the PD brain, using a delivery pump device implanted into patients’ abdomen.  Although the results were inconsistent, due to the difficulty of delivering protein continuously into the brain via an implanted pump, some patients have shown a significant clinical response to the treatment.  It is therefore not a question whether this approach works, because it definitely did in some patients, but rather how it can be done more consistently.  AAV-GDNF gene therapy treatment would result in continues delivery of GDNF protein into brain, and is therefore likely to result in significant clinical benefit for PD patients.

 

AMT has recently started preclinical development of AAV-GDNF gene therapy that will introduce the gene coding for GDNF using recombinant adeno associated virus vector (AAV).  AAV serotype 5 has been shown to be the serotype of choice for gene delivery into the brain.  After successful proof of concept (POC) and toxicology studies in rodents and primates, AMT will start an extensive clinical development.

 

Preliminary timelines for AMT-090

 

Task

 

Timelines

POC rats

 

2010

POC non-human primates

 

2010

Toxicology study

 

2011

IND / INPD (approval for phase I)

 

2011

Start phase I

 

2012

Start phase II

 

2014

Start phase III

 

2017

Filing

 

2020

Market introduction

 

2022

 



 

Project Plan Details:

 

The aim of this project is to develop a gene therapy product for the treatment of Parkinson’s disease, and to deliver a data package that is suitable for the submission and approval by the European and North American regulatory authorities.

 

Vector development and manufacturing

 

To develop a gene therapy for Parkinson’s disease, AAV-GDNF product was designed to expresses the human GDNF and is produced in insect cells using the recombinant baculovirus method.  The AAV5-GDNF is based on AMT’s standard manufacturing process, but in addition incorporates recent new technology of the basic process and makes use of an optimized Rep baculovirus construct in the upstream process and an additional chromatography step in the downstream process.  This optimisation delivers enhanced quality and robustness of the AAV5-GDNF product.  This process is fully scalable and allows for manufacturing of sufficient GMP-compliant product for PD patients.

 

Characterization of AAV5-GDNF

 

The AAV5-GDNF was tested in a functional in vitro assay in cultured E13.5 rat DRG explants.  Vigorous neural outgrowth was observed, indicating that the produced AAV5-GDNF is capable of mediating secretion of biologically functional recombinant GDNF.

 

In vivo characterization

 

Subsequently, an in-vivo characterisation of the AAV5-GDNF has been conducted.  Three different concentrations of AAV5-GDNF were injected unilaterally into the rat striatum.  Brains were analyzed for GDNF expression 6 weeks post injection using immunohistochemistry.  Resulting data demonstrated that there is a strong, concentration dependent GDNF expression throughout the injected hemisphere.

 

PoC in pre-clinical models

 

The produced AAV5-GDNF will be used to show biological activity and efficacy in animal models of Parkinson’s disease.  These experiments will be conducted using rat models of Parkinson’s disease (in collaboration with University of Lund, Sweden) as well as non-human primates model of Parkinson’s disease (in collaboration with CEA, Paris, France).  In addition to distribution studies, onset and kinetics of GDNF expression, neurochemical measurements (dopamine and dopamine metabolites), immunohistochemistry and behavioural studies will be conducted to test for functional improvement.

 

GLP Toxicology

 

The definitive design of the actual studies will be finalized after discussions with relevant agencies.  We propose to conduct a six months study in mice and in parallel a 6-12 months study in non-human primates to account for the safety of the drug.  The studies will comprise four test groups: 1. Control (vehicle), 2. Low dose (No observed effect level (NOEL) in the proof-of concept studies), 3. Mid-dose (highest dose considered for clinical studies), and 4. High dose (10 times higher than the mid-dose).

 

The protocol will include the following evaluations:

 

·                   Clinical Signs: recorded daily, beginning 7 days prior to surgery

·                   Food Consumption: recorded daily, beginning 7 days prior to surgery

·                   Body Weight: Once pre-surgery, day of surgery, then bi-weekly

 



 

·                   Clinical Chemistry: Twice a month presurgery, one week post surgery, then monthly

·                   Hematology: Twice a month presurgery, one week post surgery, then monthly

·                   Coagulation: Twice a month presurgery, one week post surgery, then monthly

·                   Antibodies against GDNF or AAV5 in plasma, twice prior to surgery, monthly thereafter.

·                   PK - CSF: To determine if there is GDNF in the CSF, twice prior to surgery, monthly thereafter.

·                   Neurological Examination: Twice prior to surgery, Day 7 post surgery, monthly thereafter

·                   MRI (T1.T2): Once prior to surgery, within three hours post surgery, and within three days prior to necropsy.

·                   Pathology

 

1.                                       Gross pathology at necropsy

 

2.                                       Selected peripheral tissues collected for histopathological analysis by a Board Certified Pathologist

 

3.                                       Complete CNS histopathological assessment by a Board Certified Neuropathologist, peer reviewed by another Board Certified Pathologist

 

·                   Q-PCR in selected organs in order to assess any biodistribution of the vector DNA to other organs.

 

Phase I/II


The primary objective of the clinical phase I/II will be to assess the safety and feasibility of intra-putaminal delivery of AAV5-GDNF to patients with PD.  Secondary objectives include measuring clinical efficacy and demonstrating improvement in a surrogate marker end point (
18 F-Dopa PET) as proof of concept.

 

We are proposing a single centre open label trial of striatally delivered AAV5-GDNF in PD employing a dose escalation design to assess the mentioned primary and secondary outcome measures.  We anticipate enrolling 12 patients in this study, with an escalating dose group design with three patients in each dose group.  We will start with the lowest dose and progress in an incremental way to higher doses.

 

Primary outcome assessments will be performed at two weeks, one month, three months, six months, 12 months and 18 months post intra-putaminal infusion of AAV5-GDNF.  Clinical secondary outcome assessments will be performed at three months, six months, 12 months and 18 months post intra-putaminal infusion of AAV5-GDNF.  18 F-dopa PET secondary outcome assessments will be performed at six months and 18 months post intra-putaminal infusion of AAV5-GDNF.

 

If feasibility and safety is confirmed and, serial PET imaging demonstrates increased 18 F-dopa uptake with a trend towards clinical improvement, we will proceed to phase 2/3 clinical trials.

 

Phase lI/III. Phase III & Regulatory submission

 

After successful completion of Phase I/II study, two additional clinical trials will be required.  The final plans for these trials will be optimised based on the outcome of the Phase I/II study.  We estimate 50 patients to be enrolled in the Phase lI/IIl clinical study, and 500 patients to be enrolled in the pivotal trial, the details however will be established, based on the outcome of the Phase I/II trial.

 



 

EXHIBIT A - Request for Benchmark Exemption With Institut Pasteur

 

Walenta, Jeffrey (NIH/OD) [E]

 

From:

Tamara Tugal [ ttugal@amtbiopharma.com]

Sent:

Tuesday, October 19, 2010 11:36 AM

To:

Walenta, Jeffrey (NIH/OD) [E]

Cc:

Mark Chadwick

Subject:

Manufacture for not-for-profit organisations and academic institutions exemption; L-107-2007 and L-119-207

 

Dear Jeffrey

 

It was nice to talk to you the other day and to have the opportunity to discuss with you AMTs plans to participate in the development of products for the treatment of ultra-orphan disorders.  We appreciate your openness to help in the development of products for rare disorders that are being developed by not-for-profit organisations.  I know that Mark is working with you on a separate amendment to the license, but I would like to take care of this particular point separately.

 

Sanfilippo Syndrome IIIB (Sanfilippo B) is a lysosomal storage disorder caused by a deficiency of the enzyme a -N-acetylglucosaminidase (NaGlu), resulting in a severe degenerative pathology of central nervous system.  Sanfilippo B patients appear normal at birth but develop hyperactivity, sleep disorders, loss of speech, mental retardation and dementia in early childhood.  Patients with Sanfilippo B will die at around 10-15 years of age.  No treatment or cure is currently available.  However, only estimated 20 children are born annually with the disease in Western Europe.  These numbers are far too small to justify commercial investment in the therapy.  Institut Pasteur has limited charity funding available for the development of treatment for this disease, and would like AMT to manufacture the clinical material for them.  AMT would like to clarify our position in relation to NIH while we manufacture to Pasteur, as we discussed on the phone.  Please see below the proposed text for such an amendment, I hope that it will be acceptable.  Please note that in addition to the L-107-2007 license, the L-119-207 license in also applicable, and we would like to have the same arrangement under both agreements to allow us to work with not-for-profit organisations.

 

Note that we have been approached by Universities attempting to develop treatments for similar diseases.  Hence we would like to be able to extend the mechanism to additional product it the future.

 

Proposed Amendment: Manufacture for not-for-profit organisations and academic institutions exemption.

 

AMT will be free to manufacture clinical trial material using the licensed technology for not-for profit organisations and academic institutions without any obligation of payments to NIH.  In the first instance, AMT intends to manufacture material for Institut Pasteur for the treatment of Sanfilippo B syndrome.  AMT will have an option to license the program from Institut Pasteur and may acquire the program from Institut Pasteur in the future.  If AMT acquires the program

 



 

from Institut Pasteur, it will, from this point onwards, pay NIH the milestones and royalties as defined in the L-107-2007 (and / or the L-119-207) license.  The mechanism will then be applicable to manufacturing of other products for not-for-profit and academic organisations.  AMT will notify NIH of any new products that it intends to manufacture under this exemption, prior to the commencement of manufacturing.

 



 

Exhibit B - PHS Consent for Benchmark Exemption with Institut Pasteur

 

 

(301) 435-5378

 

March 4, 2011

 

Tamara Tugal
Business Development Manager
Amsterdam Molecular Therapeutics
Mcibergdreef 61
1105 BA Amsterdam
The Netherlands
Phone Number: +31(0)20 566 7509

 

Re:                              Benchmark Exemption for a Proposed Collaboration Between Amsterdam
Molecular Therapeutics (‘AMT’) and Institut Pasteur

 

Dear Ms. Tugal:

 

Thank you for your request dated October 19, 2010 for a benchmark royalty exemption for collaboration agreements with not-for-profit organizations or academic institutions to conduct clinical development of potential treatments for ultra-orphan diseases.

 

Pending execution of the First Amendment to PHS License Reference No. L-107-2007/0 effective May 2, 2007 (the ‘Agreement’), PHS provides consent per Article 15 for AMT to provide material for use in clinical development of a treatment for lysosomal storage disorder Sanfilippo Syndrome IIIB at the Institut Pasteur,

 

Congratulations on your new collaboration.  When AMT makes a decision about acquisition of clinical development from Institut Pasteur, please inform PHS as soon as possible.

 

We appreciate AMT’s continued interest in PHS technologies.  If you have any questions, please do not hesitate to contact our office at any time

 

 

Sincerely,

 

 

 

/s/ Richard U. Rodriguez

 

Richard U. Rodriguez

 

Director, Division of Technology Development & Transfer

 



 

ATTACHMENT 1 - ROYALTY PAYMENT OPTIONS

 

The OTT License Number MUST appear on payments, reports and correspondence.

 

Automated Clearing House (ACH) for payments through U.S. banks only

 

The NTH encourages our licensees to submit electronic funds transfer payments through the Automated Clearing House (ACH).  Submit your ACH payment through the U.S. Treasury web site located at: https://www.pay.gov . Locate the “NIH Agency Form” through the Pay.gov “Agency List”.

 

Electronic Funds Wire Transfers

 

The following account information is provided for wire payments.  In order to process payment via Electronic Funds Wire Transfer sender MUST supply the following information within the transmission:

 

Drawn on a U.S. bank account via FEDWIRE should be sent directly to the following account;

 

Beneficiary Account:

Federal Reserve Bank of New York or TREAS NYC

Bank:

Federal Reserve Bank of New York

ABA#

021030004

Account Number:

75080031

Bank Address:

33 Liberty Street, New York, NY 10045

Payment Details:

License Number (L-XXX-XXXX) Name of Licensee

 

Drawn on a foreign bank account should be sent directly to the following account. Payment must be sent in U.S. Dollars (USD) using the following instructions:

 

Beneficiary Account:

Federal Reserve Bank of New York/ITS or FRBNY/ITS

Bank:

Citibank N.A. (New York)

SWIFT Code#

CITIUS33

Account Number:

36838868

Bank Address:

388 Greenwich Street, New York, NY 10013

Payment Details: (Line 70):

NIH 75080031
License Number (L-XXX-XXXX)
Name of Licensee

Detail of Charges (line 71a):

Charge Our

 

Checks

 

All checks should be made payable to “NIH Patent Licensing”

 



 

Checks drawn on a U.S. bank account and sent by US Postal Service should be sent directly to the following address:

 

National Institutes of Health (NIH)
P.O. Box 979071
St. Louis, MO 63197-9000

 

Checks drawn on a U.S. bank account and sent by overnight or courier should be sent to the following address:

 

US Bank
Government Lockbox SL-MO-C2GL
1005 Convention Plaza
St. Louis, MO 63101
Phone: 314-418-4087

 

Checks drawn on a foreign bank account should be sent directly to the following address:

 

National Institutes of Health (NIH)
Office of Technology Transfer
Royalties Administration Unit
6011 Executive Boulevard
Suite 325, MSC 7660
Rockville, Maryland 20852

 



 

NATIONAL INSTITUTES OF HEALTH

 

SECOND AMENDMENT TO L-l 07-2007/0

 

This is the second amendment (“ Second Amendment ”) of the agreement by and between the National Institutes of Health (“ NIH ”) within the Department of Health and Human Services (“ HHS ”), and UniQure biopharma B.V. (formerly Amsterdam Molecular Therapeutics N.V. (AMT)) having an effective date of May 2, 2007 and NIH Reference Number L-107-2007/0, and having been amended for the first time on December 31, 2009 ( NIH Reference L-107-2007/1) (“ Agreement ”).  This Second Amendment , having NIH Reference Number L-107-2007/2, is made between the NIH through the Office of Technology Transfer, NIH , having an address at (6011 Executive Boulevard, Suite 325, Rockville, Maryland 20852-3804, U.S.A., and UniQure biopharma B.V. (formerly Amsterdam Molecular Therapeutics N.V. (AMT)), having an office at Meibergdreef 61, 1105 BA Amsterdam, The Netherlands (“ Licensee ”).  This Second Amendment includes, in addition to the amendments made below, 1) a Signature Page, and 2) Attachment 1 (Royalty Payment Information).

 

WHEREAS, NIH and Licensee desire that the Agreement be amended a second time as set forth below in order to

 

a)                                      Change the name of Licensee from Amsterdam Molecular Therapeutics N.V. (AMT) to UniQure biopharma B.V. (UniQure).  This name change is the result of a transaction that took place on 30 March 2012, whereby AMT, a public company, was liquidated and all its operations and stocks were transferred to UniQure, a privately held company.

 

b)                                      Modify language related to financial terms associated with sublicensing, so as to cause a reduction in financial obligations due to NIH from sublicensing of the Agreement by Licensee in order to expedite the development of therapeutics for rare diseases.

 

NOW, THEREFORE, in consideration of the mutual covenants and promises contained herein, NIH and Licensee , intending to be bound, hereby mutually agree to the following:

 

1)                                      a)                                      In Cover page following the list of “licensed patent and patent application”, the name of Licensee has been changed to UniQure biopharma B.V.

 

b)                                      In the signature page under “signature of authorized official”, under “Official and Mailing Address for Agreement notices”, and under “Official and Mailing Address for Financial notices” “Amsterdam Molecular Therapeutics” has been changed to UniQure biopharma B.V.

 

c)                                       In the caption of the Agreement AMT is changed to UniQure.

 

2)                                      a)                                      Paragraph 6.6 is deleted in its entirety and replaced with the following:

 

6.6                                No multiple royalties shall be payable if any Licensed Products or Licensed Processes are covered by more than one of the Licensed Patent Rights .  In the event that this Agreement and NIH license L-116-2011/0 as amended from time to time apply to the same product sold by the Licensee or its sublicensees, then the Licensee shall only pay earned

 



 

royalties, benchmark royalties, and sublicensing royalties under NIH license L-l16-2011/0.

 

b)                                      In Appendix C, the second occurrence of Roman numeral “IV” at the bottom of the page is replaced with Roman numeral “VI”.  Section VI has been deleted in its entirety and replaced with the following:

 

Licensee agrees to pay NIH additional sublicensing royalties on the fair market value of any consideration received for granting each sublicense within sixty (60) days of the execution of each sublicense as follows:

 

(i) For any sublicense executed by the Licensee before the completion of any phase I clinical trial or foreign equivalent for any disease indication, Licensee agrees to pay a sublicensing royalty of ten percent (10%); and

 

(ii) For any sublicense executed by the Licensee after the completion of any phase I clinical trial or foreign equivalent but before the completion of any phase II clinical trial or foreign equivalent for any disease indications, Licensee agrees to pay a sublicensing royalty of six percent (6%); and

 

(iii) For any sublicense executed by the Licensee either after the completion of any phase III clinical trial or foreign equivalent for any disease of Orphan Indication or Ultra-Orphan Indication (as defined in L-l16-2011/0), or after the completion of any phase III clinical trial or foreign equivalent for any other disease indications, Licensee agrees to pay a sublicensing royalty of three percent (3%).

 

Contractual payments made by a sublicensee to the Licensee or an Affiliate received after the effective date of this Agreement for costs, services and expenses for the Licensee or Affiliate to perform research and development activities, or to conduct, supervise or participate in one or more clinical trial(s) for the development of the Licensed Products , or to manufacture clinical and commercial batches of Licensed Products , shall not be accounted for in the calculation of sublicensing royalties.

 

3)                                      Licensee shall pay NIH an amendment issue royalty in the sum of two hundred and fifty thousand US Dollars ($250,000.00) as follows:

 

i)                                          One hundred and fifty thousand Dollars ($150,000) shall be paid by Licensee within sixty (60) days of the effective date of this Second Amendment .

 

ii)                                       The remaining amount of one hundred thousand Dollars ($100,000) shall be paid to NIH upon execution by Licensee of any new sublicensing or partnership agreement, or on the first anniversary of this Second Amendment , whichever occurs first.

 



 

4)                                      In the event any provision(s) of the Agreement is/are inconsistent with Attachment 1, such provision(s) is/are hereby amended to the extent required to avoid such inconsistency and to give effect to payment information in such Attachment 1.

 

5)                                      All terms and conditions of the Agreement not herein amended remain binding and in effect.

 

6)                                      The terms and conditions of this Second Amendment shall, at NIH ’ sole option, be considered by NIH to be withdrawn from Licensee’s consideration and the terms and conditions of this Second Amendment , and the Second Amendment itself, to be null and void, unless this Second Amendment is executed by Licensee and a fully executed original is received by NIH within sixty (60) days from the date of NIH signature found at the Signature Page.

 

7)                                      This Second Amendment is effective on May 31, 2013   upon execution by all parties.

 



 

SECOND AMENDMENT TO L-107-2007/0

 

SIGNATURE PAGE

 

In Witness Whereof, the parties have executed this Second Amendment on the dates set forth below.  Any communication or notice to be given shall be forwarded to the respective addresses listed below.

 

For NIH :

 

/s/ Richard U. Rodriguez

 

5-23-13

Richard U. Rodriguez

 

Date

Director, Division of Technology Development and Transfer

 

 

Office of Technology Transfer

 

 

National Institutes of Health

 

 

 

 

 

Mailing Address or E-mail Address for Agreement notices and reports:

 

 

 

Chief, Monitoring & Enforcement Branch, DTDT

 

 

Office of Technology Transfer

 

 

National Institutes of Health

 

 

6011 Executive Boulevard, Suite 325

 

 

Rockville, Maryland 20852-3804 U.S.A.

 

 

 

 

 

E-mail: LicenseNotices_Reports@mail.nih.gov

 

 

 

For Licensee (Upon information and belief, the undersigned expressly certifies or affirms that the contents of any statements of Licensee made or referred to in this document are truthful and accurate.):

 

/s/ Jorn Alday

 

5-31-13

Jorn Alday, CEO, uniQure biopharma B.V.

 

Date

 

 

 

I.

Official and Mailing Address for Agreement notices:

 

 

 

Chief Executive Officer;

 

 

 

Legal@uniqure.com

 

 

 

 

 

 

II.

For invoices, payments, and Financial notices (including royalty payments):

 

Finance Dept

 

 

 

Finance@uniqure.com

 

 

 

 

 

 

uniQure biopharma B.V.

 

 

Meibergdreef 61

 

 

1105BA Amsterdam

 

 

The Netherlands

 

 

 

 

 

Phone:

0031 205667394

 

 

 

 

 

Fax:

0031 20 566 9272

 

 

 



 

Any false or misleading statements made, presented, or submitted to the Government , including any relevant omissions, under this Agreement and during the course of negotiation of this Agreement are subject to all applicable civil and criminal statutes including Federal statutes 31 U.S.C. §§3801-3812 (civil liability) and 18 U.S.C. §1001 (criminal liability including fine(s) or imprisonment).

 



 

ATTACHMENT 1 - ROYALTY PAYMENT OPTIONS

 

The OTT License Number MUST appear on payments, reports and correspondence.

 

Automated Clearing House (ACH) for payments through U.S. banks only

 

The NIH encourages our licensees to submit electronic funds transfer payments through the Automated Clearing House (ACH).  Submit your ACH payment through the U.S. Treasury web site located at : https://www.pay.gov .  Locate the “NIH Agency Form” through the Pay.gov “Agency List”.

 

Electronic Funds Wire Transfers

 

The following account information is provided for wire payments.  In order to process payment via Electronic Funds Wire Transfer sender MUST supply the following information within the transmission:

 

Drawn on a U.S. bank account via FEDW1RE should be sent directly to the following account:

 

Beneficiary Account:

Federal Reserve Bank of New York or TREAS NYC

 

 

Bank:

Federal Reserve Bank of New York

 

 

ABA#

021030004

 

 

Account Number:

75080031

 

 

Bank Address:

33 Liberty Street, New York, NY 10045

 

 

Payment Details:

License Number (L-XXX-XXXX)
Name of Licensee

 

Drawn on a foreign bank account should be sent directly to the following account.  Payment must be sent in U.S. Dollars (USD) using the following instructions:

 

Beneficiary Account:

Federal Reserve Bank of New York/ITS or FRBNY/ITS

 

 

Bank:

Citibank N.A. (New York)

 

 

SWIFT Code:

CITIUS33

 

 

Account Number:

36838868

 



 

Bank Address:

388 Greenwich Street, New York, NY 10013

 

 

Payment Details (Line 70):

NIH 75080031

 

 

 

License Number (L-XXX-XXXX)

 

 

 

Name of Licensee

 

 

Detail of Charges (line 71 a):

Charge Our

 



 

Checks

 

All checks should be made payable to “NIH Patent Licensing”

 

Checks drawn on a U.S. bank account and sent by US Postal Service should be sent directly to the following address:

 

National Institutes of Health (NIH)
P.O. Box 979071
St. Louis, MO 63197-9000

 

Checks drawn on a U.S. bank account and sent by overnight or courier should be sent to the following address:

 

US Bank
Government Lockbox SL-MO-C2GL
1005 Convention Plaza
St. Louis, MO 63101
Phone: 314-418-4087

 

Checks drawn on a foreign bank account should be sent directly to the following address:

 

National Institutes of Health (NIH)
Office of Technology Transfer
Royalties Administration Unit
6011 Executive Boulevard
Suite 325, MSC 7660
Rockville, Maryland 20852

 



 

NATIONAL INSTITUTES OF HEALTH

 

THIRD AMENDMENT TO L-107-2007/0

 

This is the third amendment (“ Third Amendment ”) of the agreement by and between the National Institutes of Health (“ NIH ”) within the Department of Health and Human Services (“ HHS ”), and UniQure biopharma B.V. (formerly Amsterdam Molecular Therapeutics (AMT) B.V.) having an effective date of May 2, 2007 as amended for the first time on December 31, 2009, and amended for the second time on May 31, 2013, and having NIH Reference Number L-107-2007/0, L-107-2007/1, and L-107-2007/2 respectively (“ Agreement ”). This Third Amendment , having NIH Reference Number L-107-2007/3, is made between the NIH through the Office of Technology Transfer, NIH , having an address at 6011 Executive Boulevard, Suite 325, Rockville, Maryland 20852-3804, U.S.A., and UniQure biopharma B.V. (formerly Amsterdam Molecular Therapeutics (AMT) B.V.), having an office at Meibergdreef 61, 1105 BA Amsterdam, The Netherlands (“ Licensee ”). This Third Amendment includes, in addition to the amendments made below, a Signature Page.

 

WHEREAS, NIH and Licensee desire that the Agreement be amended a third time as set forth below in order to a) clarify the Field of Use , and b) to update appendices D and E of the Agreement to capture all of Licensee’s past, current and future Commercial Development Plan ,

 

NOW, THEREFORE, in consideration of the mutual covenants and promises contained herein, NIH and Licensee , intending to be bound, hereby mutually agree to the following:

 

1)              In Appendix B Paragraph I, replace the Licensed Field of Use with the following:

 

Use of Licensed Patent Rights for development and sale of AAV related products

 

2)              Replace Appendix D with Appendix D attached to this Second Amendment as EXHIBIT 1.

 

3)              Replace Appendix E with Appendix E attached to this Second Amendment as EXHIBIT 2.

 

4)              All terms and conditions of the Agreement not herein amended remain binding and in effect.

 

5)              The terms and conditions of this Third Amendment shall, at NIH sole option, be considered by NIH to be withdrawn from Licensee ’s consideration and the terms and conditions of this Third Amendment , and the Third Amendment itself, to be null and void, unless this Third Amendment is executed by Licensee and a fully executed original is received by NIH within sixty (60) days from the date of NIH signature found at the Signature Page.

 

6)              This Third Amendment is effective on the date of execution by the last party to execute this Third Amendment .

 



 

THIRD AMENDMENT TO L-107-2007/0

 

SIGNATURE PAGE

 

In Witness Whereof, the parties have executed this Third Amendment on the dates set forth below. Any communication or notice to be given shall be forwarded to the respective addresses listed below.

 

For NIH :

 

 

 

 

 

/s/ Ricahrd U. Rodriguez

 

11-6-13

Richard U. Rodriguez

 

Date

Director, Division of Technology Development and Transfer

 

 

Office of Technology Transfer

 

 

National Institutes of Health

 

 

 

 

 

Mailing Address or E-mail Address for Agreement notices and reports:

 

 

 

Chief, Monitoring & Enforcement Branch, DTDT

 

 

Office of Technology Transfer

 

 

National Institutes of Health

 

 

6011 Executive Boulevard, Suite 325

 

 

Rockville, Maryland 20852-3804 U.S.A.

 

 

 

 

 

E-mail: LicenseNotices_Reports@mail.nih.gov

 

 

 

For Licensee (Upon information and belief, the undersigned expressly certifies or affirms that the contents of any statements of Licensee made or referred to in this document are truthful and accurate.):

 

/s/ Piers J. Morgan — PJ Morgan CFO

 

November 11, 2013

Piers J Morgan, CFO, uniQure biopharma B.V.

 

Date

 

 

 

I

Official and Mailing Address for Agreement notices:

 

Chief Executive Officer;

 

 

 

Legal@uniqure.com

 

 

 

 

 

 

II

For invoices, payments, and Financial notices (including royalty payments):

 

Finance Dept

 

 

 

Finance@uniqure.com

 

 

 

 

 

uniQure biopharma B.V.

 

 

Meibergdreef 61

 

 

1105BA Amseterdam

 

 

The Netherlands

 

 

 

 

 

Phone:

0031 205667394

 

 

Fax:

0031 20 566 9272

 

 

 



 

Any false or misleading statements made, presented, or submitted to the Government , including any relevant omissions, under this Agreement and during the course of negotiation of this Agreement are subject to all applicable civil and criminal statutes including Federal statutes 31 U.S.C. §§3801-3812 (civil liability) and 18 U.S.C. §1001 (criminal liability including fine(s) or imprisonment).

 



 

EXHIBIT 1

 

APPENDIX D — BENCHMARKS AND PERFORMANCE (L-107/2007)

 

Licensee agrees to the following Benchmarks for its performance under this Agreement and, within thirty (30) days after achieving a Benchmark , shall notify PHS that the Benchmark has been achieved.

 

Note:                   No formal Phase III clinical trial is required for Marketing Approval for any Orphan Indication

 

Benchmarks for lead Licensed Product (AMT-011 also known as Glybera™)

 

I.                                         Initiation of Preclinical Development phase or foreign equivalent — 4 th  quarter 2006

II.                                    Initiation of first Phase I clinical trial or foreign equivalent — 3 rd  quarter 2007

III.                               Initiation of first Phase II clinical trial or foreign equivalent — 3 rd  quarter 2007

IV.                                Submission to Regulatory Authority of first Marketing Approval or foreign equivalent — 1 st  quarter 2010

 

Benchmarks for another Orphan Indication Licensed Product (AMT-021 or equivalent)

 

I.                                         Initiation of Preclinical Development phase or foreign equivalent — 4 th  quarter 2010

II.                                    Initiation of first Phase I clinical trial or foreign equivalent — 4 th  quarter 2012

III.                               Initiation of first Phase II clinical trial or foreign equivalent — 4 th  quarter 2016

IV.                                Submission to Regulatory Authority of first Marketing Approval or foreign equivalent — 2 nd  quarter 2018

 

Benchmarks for a non- Orphan Indication Licensed Product (AMT-090 or equivalent)

 

I.                                         Initiation of Preclinical Development phase or foreign equivalent — 3 rd  quarter 2011

II.                                    Initiation of first Phase I clinical trial or foreign equivalent — 2 nd  quarter 2013

III.                               Initiation of first Phase II clinical trial or foreign equivalent — 2 nd  quarter 2017

IV.                                Initiation of first Phase III clinical trial or foreign equivalent — 4 th  quarter 2020

V.                                     Submission to Regulatory Authority of first Marketing Approval or foreign equivalent — 2024

 



 

EXHIBIT 2

 

APPENDIX E — COMMERCIAL DEVELOPMENT PLAN (L-107-2007)

 

The table below (table 1) presents a comprehensive list of all uniQure research and development projects utilizing the Licensed Patent Rights, according to main disease site and divided into projects that are, a) commercial projects, b) already in development stages, c) active research (there is already internal research activity ongoing and d) exploratory research projects (currently being considered as potential projects worth further investigation in the near future).

 

Table 1: uniQure R&D projects

 

 

 

 

 

Brain & CNS
(AAV5 based unless otherwise indicated)

Commercial
Projects

 

·                   Lipoprotein Lipase Deficiency (Glybera)

(AAV1 based)

 

 

 

Liver

(AAV5 based unless otherwise indicated)

 

 

Development Projects

 

·                   Acute Intermittent Porphyria (AMT-021)

 

·                   Hemophilia B (AMT-060)

 

 

·                   Sanfilippo B (MPS IIIB)
(AMT-110)

 

·                   Parkinson’s Disease (AMT-090) (AAV2 based)

 

Active Research Projects

 

·                   Hemophilia A (HA)

 

·                   Cirrhosis

 

·                   Hyperoxaluria (HP1)

 

·                   Huntington’s Disease

 

·                   Multiple System Atrophy (MSA)

 

·                   Hearing loss

 

Exploratory Research Projects

 

·                   Phenylketonuria (PH1)

 

·                   Ornithine transcarbamylase deficiency (OTCD)

 

·                   Glycogen storage disease type II (Pompe)

 

·                   Maroteaux-Lamy syndrome (MPS VI)

 

·                   Fabry disease

 

·                   Amyotrophic lateral sclerosis (ALS - motor neurone disease or Lou Gehrig’s disease)

 

·                   Spinal muscular atrophy (SMA)

 

·                   Batten’s disease

 

·                   Tay Sacks

 

·                   Krabbe disease (globoid cell leukodystrophy or galactosylceramide lipidosis)

 

Detailed information on the commercial, development and active research projects is provided below.

 

NOTE:  All dates contained in this Commercial Development Plan are projected estimates only.

 

A)  Commercial Projects

 

Glybera

 

Glybera, an AAV1 based product for lipoprotein lipase deficiency, was approved under exceptional circumstances by the EMA in October 2012. Currently uniQure is in the phase of preparation of product launch, for which it has

 



 

found a partner in Chiesi. It is our aim together with Chiesi to launch in Europe in the first half of 2014. The commercialization agreement with Chiesi has been shared with NIH.

 

uniQure is actively working on post-approval commitments which came with the approval under exceptional circumstances. Besides some CMC related commitment, there is a commitment for a Phase IV study to collect more biomarker data on the chylomicron handling before and after treatment with Glybera as well as setting up a registry for LPLD patients. Both studies are in progress and in accordance with timelines of the commitments to the EMA.

 

Furthermore, uniQure has initiated the first campaign for the product of commercial product. This campaign is ongoing and will result in release of the first batch in quarter 1, meeting the planning of the launch of Glybera.

 

In the context of the European pricing and reimbursement (P&MA) preparations for Glybera, these have been initiated and efforts are ongoing to prepare a pricing strategy as well as a European Core value Dossier (CVD). The agency selected to support the development of the pricing strategy as well as the European CVD has been nominated following a thorough screening and selection process, - the mutual CDA does not allow a disclosure of the agency, which has been agreed with our partner Chiesi. Please be aware that the continued management of the P&MA preparations is now led by Chiesi, who have the commercialization rights for Glybera in the EU.

 

The P&MA dossier is the crystallization of a multifactorial approach to P&MA and will focus on the Clinical and Epidemiology Data & Publications, Economic Tools & Data, P&MA Strategy and Goals as well as P&MA Implementation & Tactics.

 

The Initial evaluation and assessment of the current available market information is sparse due to the new concept of introducing the first gene therapy, a market which today has basically no treatment for LPLD patients and the first time introduction of a one-time administration treatment for an ultra-rare disease.

 

Key clinical data will be incorporated into the CVD and translated into value statements and economic benefits that will provide rationale and evidence for the positioning of Glybera® with key stakeholders, especially physicians and payers.

 

The Core Value Dossier is, therefore, a crucial vehicle to deliver the P&MA strategy for Glybera® and support tactical execution. Based on the initial preparations, a stepwise approach to the design and development of the Core Value Dossier for Glybera® has been planned.

 

In the event that serious gaps exist in the data and/or major problems are identified in this initial review, with regard to the usability and credibility of the data with payers and customers, these issues will be discussed between Chiesi and uniQure.

 

The Glybera® Core Value Dossier will be the internal reference document summarizing the available evidence and documentation and developing these into value statements and approaches to the various relevant stakeholders.

 

The CVD structure is designed to bring consistency to the messages delivered in each country and, at the same time, provide the depth of information on each point to allow for the essential local market tailoring.

 

As per 30th September 2013, Chiesi has reported having one informal meeting with a European Health authority to discuss the process and management of the P&MA dossier.  As the meeting was informal there was no agenda and no official minutes have been taken. As per 30th September, no reimbursement reports are available. The on-going development of the CVD and the pricing strategy are managed by the same agency.

 

Discussions with the FDA were started in August of 2013, which will be followed by filing of an IND in Q1 of 2014.

 

uniQure is currently developing a geographical expansion strategy to find partners and product approval in other

 



 

territories, such as Israel, Canada and South Korea.

 



 

Liver Programs

 

B) Development Programs

 

1.               AMT-021 for Acute Intermittent Porphyria

 

Disease Background

 

Acute Intermittent Porphyria, or AIP, is a rare liver metabolic disorder resulting from mutations in the PBGD gene. This gene encodes for the enzyme porphobilinogen deaminase (also known as hydroxymethylbilane synthase — HMBS), a liver protein necessary for the production of heme, a component of hemoglobin and other blood proteins. Insufficient activity of this protein leads to an accumulation of toxic metabolites (ALA and PBG), resulting in a wide variety of serious clinical problems, including acute, severe abdominal pain, muscular weakness and an array of neurologic manifestations, including psychiatric episodes, seizures and coma. In the majority of cases, attacks are triggered by precipitating factors such as hormonal fluctuations, infections, drugs and dietary changes. Long-term consequences may include irreversible nerve damage, liver cancer and kidney failure. Patients with AIP experience regular hospitalizations and extremely poor quality of life, and may in some cases require liver transplants. Acute attacks can be life-threatening. Current therapies only target the disease symptoms and do not prevent attacks or fully minimize or control their consequences.

 

A recent epidemiological study reported that, in Europe (excluding Sweden), the incidence of AIP is 0.13 per million population per year and based on that they estimated a prevalence of 5.9 per million population (Elder et al., 2012). In Sweden the incidence and prevalence of AIP are about four times higher than in the rest of Europe due to a founder effect originating in Lappland (Floderus et al., 2002).  The frequency in the United States is estimated to be 1-5 cases per 100,000 population ( www.emedicine.medscape.com/article/205220-overview#a0199 ).

 

Overview of AMT-021 Program

 

The goal of our AMT-021 program is to provide long-term normalization of the PBGD protein in order to prevent acute AIP attacks and their complications.

 

The program has been developed through a collaborative agreement with the Foundation for Applied Medical Research (FIMA), its Center for Applied Medical Research (CIMA) and its commercialization arm, DIGNA Biotech, of the University of Navarra (Pamplona, Spain). Part of the funding to support for the Phase I trial (including GLP safety & toxicology studies and the observational trial) was secured through the European Commission Framework Programme 7  award (€3.3 million, grant agreement 261506) made to the AIPGENE consortium ( www.aipgene.org/ ), of which uniQure is a partner.

 

UniQure holds an exclusive license to the gene cassette being used in the Phase I clinical trial. Under our agreement with DIGNA Biotech and the other consortium members, Licensee has an exclusive right to all data related to the program.

 

Preclinical Development

 

·                   Product Profile

 

AMT-021 is designed to be delivered systemically through a peripheral vein in a single administration.

 

AMT-021 or rAAV5-hPBGD, is a recombinant adeno-associated vector of serotype 5, consisting of:

 



 

·                   Inverted terminal regions or ITRs of the adeno-associated serotype 2

·                   A human codon optimized porphobilinogen deaminase gene or hPBGDco as the therapeutic gene

·                   A liver specific promoter constituted by the albumin enhancer (Ealb) and the alfa-1-antitrypsin promoter (hAAT)

 

·                   Pre-clinical Proof of Concept

 

Pre-clinical proof of concept (PoC) studies have been performed using the AIP murine model developed by Lindberg et al. (1999).  In these studies, long term therapeutic efficacy was achieved.  More specifically, at 5x10 13  gc/kg, metabolic correction of the hepatic PBGD enzyme activity, normalization of the PBG and ALA precursor’s accumulation in urine and improvement of the motor coordination were observed. Additionally, a complete neurological study indicated the correction of neurotoxic porphyrin precursors was able to restore nerve conduction and the impaired peripheral neuropathy.

 

In non-human primates (NHP) treated with AMT-021 at a dose of 5x10 13  gc/kg endogenous PBGD enzymatic activity increased by a factor of two in male and between three and five times in female animals.

 

·                   Non-clinical safety & toxicology studies

 

The following table presents a summary of the AMT-021 non-clinical safety and toxicology studies that have been conducted to support the clinical development program.

 

Parameter to be
assessed

 

Study performed

 

Results

Pharmacokinetics

 

1) GLP biodistribution in C57Bl/6 mice (180 days) with validated QPCR method.

 

2) Biodistribution data in Rhesus macaques (supportive)

 

In both species,

 

·              Clear correlation between the dose of vector administered and the genome copies of the vector as well as vector RNA (expression) quantified in the liver.

·              Tissue biodistribution was mainly limited to liver although some significant transduction was detected in spleen, lymph nodes, heart and adrenal glands.

 



 

Toxicity

 

1) GLP toxicity study in Rhesus macaques, 30 days

 

2) GLP toxicity study in C57Bl/6 mice (180 days)

 

In Rhesus macaques:

 

·              No specific macroscopic or microscopic findings, no severe immune/inflammatory reactions (NOAEL 5x10 13  gc/kg)

 

In C57Bl/6 mice:

 

·              No severe adverse toxicologically significant findings

·              At highest dose limited hepatocyte vacuolation due to metabolic adaptation to high PBGD activity in liver (NOAEL 5x10 14  gc/kg)

Off-target expression

 

Included in the GLP toxicity study in monkey and mouse

 

Vector RNA analysis for all tissues other than liver showed that there is no PBGD transcription and translation elsewhere.

Shedding pattern

 

Included in the Rhesus macaques study

 

Shedding in semen and saliva is transient (cleared by day 30)

Persistence in semen and risk of germline transmission

 

Mouse Germline transmission study in C57Bl/6 mice

 

No transmission to offspring

Carcinogenicity

 

Analyze tissues from the Rhesus studies using LAM-PCR

 

·              Low frequency of integration sites, with random distribution

·              No preferences for CpG islands

·              No preference for palindromic regions

·              No hotspots (low clustering level)

 

1.1.1                                                                                              Summary of AMT-021 Preclinical Development Program

 

Single intravenous administration of AMT-021 into wild type mice and Rhesus macaques results in:

 



 

·                   Efficient liver transduction resulting in dose dependent increase in viral RNA copy numbers and in turn producing increased PBGD activity

 

·                   No morbidity, no changes in body weight or food intake

 

·                   No changes in biochemistry, hematology, coagulation and urinalysis associated with AAV5-hPBGD

 

·                   Negative vector shedding 30 days after viral administration in serum, saliva, nasal secretions, urine, faeces and semen

 

·                   Tissue biodistribution that is mainly limited to liver although some significant transduction was detected in spleen, lymph nodes, heart and adrenal glands

 

·                   Specific hepatic PBGD expression

 

Clinical Development Program

 

The key regulatory and clinical development best estimate milestones for AMT-021 include the following,

 

·                   EMA Orphan Drug Designation (EU/3/09/632)

Apr 2009

 

 

·                   FIMA/ CITA/ UTE/ DIGNA - AMT Collaborative Agreement

May 2010

 

 

·                   EU-FP7 AIPGene Consortium

Jan 2011

 

 

·                   Observational Study AEMPS approval

May 2011

 

 

·                   Observational Study start

Jul 2011

 

 

·                   Phase I Study AEMPS approval

Oct 2012

 

 

·                   Phase I Study: first patient treated

Dec 2012

 

 

·                   Phase I Study: last patient treated

Aug 2013

 

 

Expected milestones

 

 

 

·                   Phase II/III start:

4Q2016

 

 

·                   MAA/ NDA submission:

2Q2018

 

 

·                   Observational trial

 

 

A prospective non-interventional (pre-treatment) observational study started at the end of 2011 that aims to assess the evolution of disease-related clinical and laboratory parameters in time, as well as characterize aspects of disease management such as AIP-related hospitalization. This baseline assessment is intended to study possible relationships

 



 

between biochemical parameters and clinical endpoints that will in turn be valuable in evaluating any signs of efficacy in the Phase I trial as well as in subsequent trials. Eight patients are expected to be enrolled who after completion of this observational phase would then enter the interventional stage of the program, i.e., first-in-human clinical study (Phase I). The observational study is to last for at least six months for each participant.

 

To date all 8 AIP-patients have been recruited into the observational study and all but one have completed a minimum of 6 months pre-treatment assessments.  The last patient completed the observational study in August 2013.

 

·                   Phase I trial

 

The Investigational Medicinal Product Dossier (IMPD) was submitted to the AEMPS (Spanish Agency for Medicines and Medical Devices) in June 2012 and was approved by the Agency in October 2012.

 

The Phase I study is a multicenter, open label, prospective, interventional, single dose, dose-escalation clinical trial to investigate the safety and tolerability of AAV5-hPBGDco (AMT-021) in patients with severe Acute Intermitted Prophyria (Eudra CT no. 2011-005590-23).

 

The primary objective is to assess the safety of systemic administration and determine the maximum tolerated doses (MTD). Secondary objectives include urinary levels of toxic metabolites (ALA and PBG), disease symptoms evaluation, quality of life evaluation and assessment of pharmacokinetics. Exploratory objectives include, neurological involvement, identification of novel biomarkers and pharmacokinetic modeling.

 

The Phase I study was initiated in December 2012 in the Department of Medicine (Liver Unit) at the University Clinic of the University of Navarra (Pamplona, Spain). There are two patients per cohort and four cohorts in the trial (each cohort receiving 5x10 11 , 2x10 12 , 6x10 12  or 1.8x10 13  gc/kg) and all patients will be followed- up for one year as part of the Phase I study.

 

All 8 patients who completed the observational trial have also been treated as part of the Phase I study.  In the 8 treated patients, no Serious Adverse Events, Treatment Emergent Adverse Events or Liver Events (Dose Limiting Toxicities - DLT’s) related to the study medication have been observed to date.

 

·                   Future Clinical Development

 

It is envisaged that the Phase II/III will be a confirmatory trial where the study population and the outcomes to be assessed (efficacy endpoints — clinical and biochemical) will be based on those as for Phase I. Licensee also intends to carry out the study in both Europe and the USA .

 

Summary of AMT-021 Clinical Development Program

 

·                   The first time an AAV5 gene therapy product has been tested in humans

 

·                   The first time an AAV gene therapy product has been tested in humans at such high dose, i.e., 1.8x10 13  gc/kg

 

·                   No Serious Adverse Events, Treatment Emergent Adverse Events or Liver Events (DLT’s) related to the study medication have been observed in the Phase I study to date

 



 

·                   The Phase I is expected to be completed in 3Q14 and Phase II/III is expected to start by the end of 2014

 

·                   The Phase II/III program will run in parallel in Europe and US where MAA and NDA, respectively, are expected in 4Q2016

 

2.               AMT-060 for Hemophilia B

 

Disease Background

 

Hemophilia B is a serious inherited orphan disease in males characterized by insufficient blood clotting. The condition can lead to repeated and sometimes life-threatening episodes of external and internal bleeding following accidental trauma or medical interventions. The episodes may cause long-term damage, for example to the joints, and may be fatal if they occur in the brain. The deficient blood clotting is caused by the lack of functional human Factor IX, or hFIX, a blood clotting factor, as a result of mutations in the gene responsible for encoding this essential protein. The presence of hFIX at greater than 1% of normal levels has a therapeutic effect in promoting clotting. The current standard treatment is prophylactic protein replacement therapy, in which frequent intravenous administrations of recombinant Factor IX (often 2-3 times per week) are required to stop or prevent bleeding. Protein replacement therapy is costly ($150,000-200,000 per patient per year) and burdensome, and does not completely prevent bleeding.

 

The total Hemophilia B patient population in the European Union and the United States is estimated at approximately 25,000, according to the World Federation of Hemophilia 2010 Report on the Annual Global Survey. About 40% of individuals with the disease have a severe disorder, characterized by functional factor IX levels that are less than 1% of normal, whereas moderately severe Hemophiliacs (about 30% of the Hemophiliac population) have 1%-5% of normal and those with the mild phenotype (the remaining 30%) have between 5% and 40% of normal factor IX levels ( www.orpha.net ). Based on these estimates Licensee believes that approximately 70-85% of the worldwide patient population would be eligible for treatment with gene therapy. Licensee believes that the treatment would not be appropriate for those patients with very mild disease phenotype.

 

Overview of AMT-060 Program

 

The goal of our AMT-060 program is to restore blood clotting on a long-term basis through the introduction of the functional gene for hFIX into the patient’s liver cells.  Licensee is currently in the process of finalizing pivotal (GLP) safety and toxicology studies and preparing to conduct a Phase I trial.

 

Preclinical Development

 

·                   Product Profile

 

AMT-060 is designed to be delivered systemically through a peripheral vein in a single administration .

 

The use of recombinant adeno-associate vectors (rAAV) of serotype 5 (rAAV5) for targeted gene delivery to the liver was pioneered by St. Jude Children’s Research Hospital (SJCRH) where for pre-clinical experiments the hFIX expression cassette was packaged into AAV5 capsids in HEK-293T mammalian cells. HEK-293 produced AAV5-hFIX is not suitable for further development because as a production system it is not amenable to large-scale production. To allow up scaling, the expression cassette has now been transferred into uniQure’s proprietary baculovirus expression vector system (BEVS) that can be adapted to a GMP setting. The resulting vector produced

 



 

using the baculovirus expression system is termed AAV5-hFIXco or AMT-060.  Licensee also holds a license from SJCRH to the gene cassette used in the currently ongoing Phase I/II AAV 2/8-LP1-hFIXco trial.

 

AMT-060, rAAV5-hFIXco, is a recombinant adeno-associated vector of serotype 5, consisting of:

 

·                   Inverted terminal regions (or ITRs) of the adeno-associated serotype 2

·                   A human codon optimized FIX gene (or hFIXco) as the therapeutic gene

·                   The liver specific promoter, LP1, derived from the human apolipoprotein hepatic control region and the human alpha-1-antitrypsin (or hAAT) promoter

 

·                   Virus serotype selection

 

The hFIXco expression cassette and rAAV5 or AAV8 vectors have been extensively studied in mice and non-human primate.  Both vectors have been shown to have similar tropism to (preference to transduce) the liver (Nathwani et al., 2007) and AAV5-hFIXco was shown to mediate expression of significant levels of human factor IX in non-human primates (NHP) during a monitoring period of more than 5 years (Nathwani et al., 2011). In this study none of the animals presented elevated liver enzymes levels or other signs of toxicity during the whole observation period. Liver examination by MRI scanning did not reveal any abnormalities in any of the animals.

 

These pre-clinical data suggest that i.v. administration of the AAV5-hFIXco vector is able to mediate a similar level of human factor IX as presented for AAV8-hFIXco, and such administration is not associated with safety concerns or immunogenicity against the human factor IX.

 

·                   Pre-clinical Proof of Concept

 

Pre-clinical PoC studies have been carried out in wild type mice, non-human primates (NHP) and are currently being completed in transgenic Hemophilia B mice. In wild type mice (C57Bl/6) intravenous administration of AMT-060 mice resulted in dose-dependent levels of (human) factor IX levels in murine plasma as determined by ELISA. Human factor IX levels amounted up to 11% of those in normal human plasma 4 weeks after infusion of 5x10 12  gc/kg, demonstrating that AAV5-hFIXco produced in the BEVS is biologically active.

 

In Rhesus monkeys dosed with AMT-060 (5x10 12  gc/kg) by intravenous infusion, human FIX levels peaked to 7%-16% of normal human levels one week after infusion, and stabilized to 5-10% of normal human levels 4 weeks after infusion until sacrifice (12 weeks after dosing). These kinetics are in accordance with those observed in previous studies (Nathwani et al., 2007; Jiang et al., 2006), indicating that i.v. administration of AAV5-hFIXco produced in BEVS results in a level of factor IX in plasma that is similar to that produced using AAV5-hFIXco produced in HEK293 cells. Post mortem, (RT)-QPCR demonstrated homogeneous vector DNA delivery and transgene expression in the liver. No signs of adverse reactions were observed. Infusion was associated with slight and transient effects in plasma chemistry shortly after dosing, such as a brief increase of liver enzyme activity levels, consistent with infusion of a viral protein. Necropsy revealed no significant macroscopic or microscopic abnormalities.

 

Preliminary data in Hemophilia B mice indicate that treatment with AMT-060 induces normalization of FIX levels as well as clotting time.

 

·                   Non-clinical safety & toxicology studies

 



 

The following table presents a summary of the AMT-060 non-clinical safety and toxicology studies that are being conducted to support the clinical development program.

 

Parameter to be assessed

 

Study performed

 

Status

Pharmacokinetics

 

1) GLP biodistribution in C57Bl/6 mice (180 days) with validated QPCR method.

 

2) Biodistribution data in Rhesus macaques (supportive)

 

Ongoing

Toxicity

 

1) GLP toxicity study in Cynomolgus monkeys (180 days)

 

2) GLP toxicity study in C57Bl/6 mice (180 days)

 

3) Liver pathology in Rhesus macaques (supportive)

 

Ongoing

Off-target expression

 

Included in the GLP toxicity study in monkey and mouse

 

Ongoing

Shedding pattern

 

Partly included in the mouse GLP toxicity study.

 

Partly included in the Rhesus macaques study.

 

Ongoing

Persistence in semen and risk of germline transmission

 

1) Mouse Germline transmission study in C57Bl/6 mice

 

2) QPCR of the testis in the Rhesus macaques at 90 days (non-GLP).

 

Ongoing

Carcinogenicity

 

Analyze tissues from the Rhesus studies using LAM-PCR

 

Ongoing

Juvenile toxicity

 

Ethical approval obtained and study is being planned (to support PIP plans)

 

Ongoing

 

Summary of AMT-060 Preclinical Development Program

 

·                   AAV5-hFIXco shows similar liver tropism to AAV8-hFIXco and results in significant and long lasting increase in FIX expression.

 



 

·                   Single intravenous administration of AMT-060 into wild type mice and Rhesus macaques results in significant and long lasting hFIX levels with no noticeable adverse events and no macroscopic or microscopic findings.

 

·                   GLP safety and toxicology studies are expected to be completed in December 2013 .

 

Clinical Development Program

 

The key regulatory and clinical development milestones for AMT-060 include the following,

 

·                   EMA Orphan Drug Designation:

Nov 2011

 

 

·                   FDA Orphan Drug Designation:

Jan 2012

 

 

·                   EMA Scientific Advice:

Feb 2012

 

 

·                   EMA Phase I Protocol Advice:

Aug 2012

 

 

·                   GLP Safety & Tox Studies:

2Q2012 to 4Q2013

 

 

Expected milestones

 

 

 

·                   IMPD submission:

2Q2014

 

 

·                   Phase I start:

3Q2014

 

 

·                   Phase II/III start:

3Q2016

 

 

·                   MAA/ NDA submission:

2Q2018

 

·                   Phase I trial

 

The Phase I study will be a multicenter, open label, prospective, interventional, single dose, dose-escalation clinical trial to investigate the safety and tolerability of AAV5-hFIXco (AMT-060) in patients with severe Hemophilia B.

 

The primary objective is to assess the safety of systemic administration and determine the maximum tolerated doses (MTD). Secondary objectives include:

 

·                   To estimate the appropriate dose required to achieve stable expression of hFIX at or above 3% of normal

 

·                   To evaluate kinetics (dose-related duration and magnitude) of expression

 

·                   To assess the immune response to hFIX transgene product

 

·                   To assess the immune response to the AAV5 capsid proteins

 

·                   To assess viral shedding in various body fluids (including semen)

 

·                   To assess the occurrence of FIX inhibitors

 

·                   To evaluate coagulation parameters

 

·                   To assess need for FIX concomitant treatment

 



 

Twelve male adults patients ( > 18 year old to < 35 year old) with genetically confirmed Hemophilia B and phenotypically defined as having severe disease ( < 1% of normal plasma FIX levels) are expected to be enrolled.  Initial patient follow-up will last for 12 months as part of the Phase I trial.

 

·                   Future Clinical Development

 

It is envisaged that the Phase II/III will be a confirmatory trial where the study population and the outcomes (efficacy endpoints — clinical and biochemical) will be based on those for the Phase I. Licensee will also consider expanding the patient population to moderately severe patients and intend to carry out the study in both Europe and USA.

 

Summary of AMT-060 Clinical Development Program

 

·                   The IMPD is planned to be submitted in 2Q2014

 

·                   Phase I is planned in patients with severe Hemophilia B and is expected to start in 3Q2014

 

·                   It is the intention that in Phase II/III the patient population will expand to moderately severe Hemophilia B patients

 

·                   The Phase II/III program will run in parallel in Europe and USA where MAA and NDA, respectively, are expected in 2Q2017

 

The Hemophilia B program has been partnered with Chiesi. The co-development agreement has been shared with NIH.

 

C) Active Research Projects

 

1.               Hemophilia A

 

Disease Background : Hemophilia A (HA) is a genetic, X-linked, recessive disorder caused by production of dysfunctional or by production of insufficient amount of factor VIII (FVIII) protein, a key protein involved in the blood coagulation cascade.  Hemophilia A patients suffer from spontaneous bleeding in the large joints and soft tissue, and are at risk for intracranial hemorrhage.  Recurrent episodes of joint bleeding can lead to crippling arthropathy, particularly in severely affected patients.  HA comprises the majority of hemophilia patients (80%), with incidence of ~1:10,000 to 1:50,000 males affecting 400,000 people worldwide.

 

Numerous mutations in the FVIII gene have been described giving rise to different disease phenotypes.  Similarly to Hemophilia B (HB), individuals with less than 1% active factor are classified as having severe hemophilia, those with 1—5% active factor have moderate hemophilia, and those with mild hemophilia have between 5—40% of normal levels of active clotting factor.

 

Clinical need : HA seems an excellent candidate for gene therapy (GT) as it is a well characterized monogenic disorder.  The product of the FVIII gene is a plasma protein which is normally secreted by hepatocytes and endothelial cells but can also be expressed in other cell types, e.g., adipocytes, mycoytes or fibroblasts.  Furthermore, only modest increase >1% can markedly reduce spontaneous bleedings.  The effects of gene therapy can be readily monitored by changes in phenotype and by obtaining peripheral blood to measure FVIII antigen levels and clotting factor activity.  Currently, treatment for HA consists of infusion of either plasma-derived or

 



 

rFVIII protein for bleeding episodes.  Although, prophylactic infusion of FVIII concentrates is generally effective in alleviating bleeding episodes and subsequent joint disease, the short half-life of FVIII (~12 hours) and the high cost of purified FVIII products make life-long prophylactic treatment demanding for patients and costly.

 

Feasibility

 

Gene : The gene of factor VIII is located on the long arm of the X chromosome.  It spans over 180 kb, and as such is one of the largest genes known.  It comprises of 26 exons, which encode a polypeptide chain of 2351 amino acids including a signal peptide of 19 and a mature protein of 2332 amino acids.  It is a secreted protein.  Its primary structure, deduced from the cloned factor VIII cDNA, includes discrete domain structure: A1-a1-A2-a2-B-a3-A3-C1-C26-8.  The B domain is unique in that it exhibits no significant homology with any other known protein and can be deleted with the resulting recombinant protein displaying essentially normal survival in circulation and able to correct the bleeding tendency in HA patients.

 

Vector optimization : A rAAV5 containing HLP-hFVIIIco (promoter HLP; codon optimized hFVIII with partial B-domain deletion) has been generated in-house where the preserved B-domain consists of 225 N-terminal amino acids containing 6N-glycosilation sites. This variant has been previously shown to increase secretion of FVIII as compared to wild type or to FVIII with complete B-domain deletion and in-house work with HLP-hFVIII has shown that it produces active FVIII protein in vitro and in vivo .

 

This FVIII expression cassette size is however ~5.6 kbp, which exceeds the AAV packaging limit (4,7-4,9 kbp).  In-house molecular analysis studies of the encapsidation products showed that the 5.6kbp FVIII expression cassette is not entirely encapsidated in the AAV particle. Instead + and – DNA strands of the encapsidated molecules revealed missing 5’ ends. This is consistent with previously reported unidirectional (starting at 3’ end) packaging mechanism operating according to “head-full principia” with 4.7-4,9 kbp limit. Licensee hypothesizes that the correct template for production of FVIII was assembled in the target cell based on complementation of + and – DNA strains each of which delivering the 5’ missing end. This is to be expected since + and – strain complementation is responsible for normal rAAV episome formation.  Licensee proposes therefore to develop a product that consist of incomplete + and – DNA strains, which are assembled into complete expression cassette upon episome formation.

 

A proof of concept study has been initiated involving a number of FVIII construct and including full FVIII codon optimized gene.  The study aims to characterize the viral DNA, formation of episomes upon delivery of the expression cassette to the nucleus, resulting mRNA and FVIII protein.  The potency of the vector is currently being investigated in a number of animal models.

 

It is our aim to develop this product to clinical stage Phase I by the end of 2015. Duration of clinical development and further timelines have not been defined.

 

Development overview to IMPD:

 

 



 

Completion of vector optimization work will provide the first milestone (Go/No Go) for the project.

 

Safety Assessment : The disease and gene therapy approach are similar (or equivalent) to Hemophilia B where no major safety concerns have been described.

 

2.               Cirrhosis

 

Disease Background : Liver cirrhosis is a consequence of chronic liver disease characterized by replacement of liver tissue by fibrosis, scar tissue and regenerative nodules (resulting from regeneration of damaged tissue), leading to loss of liver function.  The four leading causes of cirrhosis and primary liver cancer in Europe include harmful alcohol consumption, viral hepatitis B, viral hepatitis C and metabolic syndromes related to overweight and obesity.  The European Association for the Study of the Liver in its 2013 report reported that approximately 29 million people in the European Union suffer from a chronic liver condition and that the incidence and prevalence of two conditions, cirrhosis and primary liver cancer, are key to understanding the burden of liver disease.  Both conditions represent the end-stage of liver pathology and thus are indicative of the associated mortality.

 

The hypothesis behind this project is that liver cirrhosis is a state of IGF-I insufficiency and low expression of IGF-I locally in the liver will revert and/ or prevent further exacerbation of cirrhosis.  A confidentiality agreement concerning this project was signed between DIGNA/ CIMA and uniQure in October 2012.

 

Pre-clinical evidence to support this hypothesis includes the following:

 

·                   Proof of Mechanism: SV40-IGF-I administration in rat cirrhotic liver models resulted in increased mRNA and protein levels of IGF and IGF-BP3 compared to control animals in the hepatocytes and levels remained constant for up to 6 months (Sobrevals et al., 2010).

 

·                   Proof of Principle: SV40-IGF-I mediated expression in rat cirrhotic liver models, induced fibrolysis through upreguation of MMPs and downregulation of TIMP-1/-2, reduced expression of pro-fiborgenic factors (TGFb, amphilregulin, PDGF, CTGF, VEGF), induced antifibrogenic and cytoprotective factors (HGF), promoted hepatocyte differentiation through upregulation of HNF4a and downeregulation of WT-1 (Sobrevals et al., 2010).

 

Treatment with recombinant IGF-I protein resulted in improved hepatic function, decreased oxidative stress/ damage, improved mitochondrial function and decreased fibrosis (Castillo-Cortazar et al., 2000; García-Fernández et al., 2005; Lorenzo-Zuniga et al., 2006).

 

·                   Proof of Concept: SV40-IGF-I-mediated expression in rat cirrhotic liver reversed fibrosis by decreasing expression of collagen I & IV and deactivation of HSC, and improved liver function through normalisation of AST, ALT, ALP, bilirubin and albumin (Sobrevals et al., 2010).

 

Clinical evidence to support disease linkage includes the following:

 

·                   In patients suffering from liver cirrhosis circulating IGF-I levels (or IGF-BP3) correlate with disease severity scores; Child-Pugh and MELD (Kratzsch et al., 2005; Khoshnood et al., 2013).

 

·                   A short course (for 4 months) of IGF-I recombinant therapy treatment increased the levels of albumin and tended to improve energy metabolism (surrogates for liver function) & the levels of serum albumin positively correlated with IGF-I/IGF-I BP3 ratio (Conchillo et al., 2005).

 

Clinical need : Transplantation is the only curative option for the disease and contraindications to transplantation include, a) co-morbidities (e.g., TB), b) over 65 years of age, c) coronary artery disease and d) tumours in previous 5

 



 

years.

 

The initial target population for IGF-I gene therapy for liver cirrhosis could/ would be those cirrhotic patients with IGF-I insufficiency (i.e., 50% of all cirrhotic patients), possibly patients with Child-Pugh A and/ or B score and with IGF-I levels below normal values.  An ODD application for this specific population may be considered.  The table below indicates the Child-Pugh scoring scheme for liver disease prognosis.

 

Points

 

Class

 

One year
survival

 

Two year
survival

 

5-6

 

A

 

100

%

85

%

7-9

 

B

 

81

%

57

%

10-15

 

C

 

45

%

35

%

 

Feasibility :

 

Gene: The IGF1 gene is located on chromosome 12 and spans 7.3 kb encoding a 70 amino acid residue protein. It contains 6 exons, 4 of which are alternatively spliced depending on tissue type and hormonal environment.  The IGF1 coding region is flanked by sequences encoding an amino-terminal peptide of at least 25 residues and a carboxyl-terminal peptide of 35 amino acids which indicates that IGF1 is synthesized as a precursor protein that undergoes proteolytic processing at both ends before being secreted.

 

Vector optimization: The IGF-1 rat version is available with CIMA and was used for proof of concept studies.  Human IGF-I vectors are being developed in-house as part of work done by the Emerging Technologies Research Group on regulated gene expression systems.

 

Animal models: A rat model is available with CIMA and has been used for proof of concept studies.  A number of other small animal models have been described (Liu et al., 2013).

 

Biomarkers: Circulating IGF-I (and other related proteins) can be monitored using commercially available methodology.  However the relevance of this to liver (local) levels of IGF-I and whether GT can deliver sufficient amounts of IGF-I that that can be readily detectable in the circulation need to be established.

 

Liver function and signs of cirrhosis can be monitored following well established standard procedures (e.g., liver enzymes, markers of fibrosis etc.).

 

The PoC obtained at CIMA will have to be repeated with uniQure’s AAV5-IGF1 vector. Licensee is at the initial stages of research aiming to initiate a Phase I clinical trial by the end of 2016.

 

Development overview to IMPD:

 

 



 

The GLP safety and toxicology studies will provide the first milestone (Go/No Go) for the project.

 

Safety Assessment : Safety studies in rat disease models (8 months) and wild type rats  (8 weeks) showed no signs of toxicity due to treatment with SV40-IGF-I (Sobrevals et al., 2010).

 

Potential toxicity concerns include tumor formation and interference with insulin/ glucose metabolism albeit both issues are unlikely as the aim of this approach would be to upregulate levels of IGF-I where they are already below normal rather than to achieve supra-physiological levels.  In addition, gene therapy vectors are likely to induce lower level of localized expression without substantial increase in serum IGF-I levels.  Regarding potential for tumorigenesis, IGF-I therapy is thought to favor hepatocellular differentiation, i.e., opposes carcinogenesis, and studies have shown that sharp decrease in IGF-I in cirrhotic liver may contribute to hepatocellular carcinoma (HCC).  In addition it is believed that it is IGF-II that is the key player in HCC.  Furthermore, patients with existing tumor nodules in their liver could/ should be excluded from trials.

 

[NOTE: Hepatocellular carcinoma occurs at a rate of 1% to 4% per year after cirrhosis is established and cirrhosis underlies HCC in approximately 80%-90% of cases worldwide (Giovanna Fattovich  et al., 2004), i.e., the vast majority of cirrhotic patients do not develop HCC or at least they do not live live long enough to develop it]

 

3.               Hyperoxaluria

 

Disease Background : Primary hyperoxaluria type I (PH1) is a rare, autosomal recessive inherited metabolic disorder characterized by a deficiency of the hepatic enzyme alanine-glyoxylate aminotransferase (AGXT), which produces a marked increase in endogenous oxalate synthesis by the liver. Oxalate is a metabolic end product in humans and excess oxalate provokes hyperoxaluria, causing progressive urolithiasis, nephrocalcinosis and chronic renal failure, ultimately leading to end-stage renal failure (ESRF) and death if untreated.

 

It is the most common and severe variant among a spectrum of metabolic disorders resulting in hyperoxaluria.  The disease has an estimated prevalence ranging from 1 to 3 per 1 million individuals and an estimated incidence of 1-9:100,000 live births per year in Europe. However, higher rates are reported in historically isolated populations, like the Canary Islands. PH1 accounts for <1% of pediatric ESRF in developed countries.

 

A pre-clinical proof of concept study has already been conducted in collaboration with Eduardo Salido (University Hospital of Canary Islands) using AGXT knockout mice demonstrating that in the GT treated animals oxalurea reduced to normal levels with restoration of liver enzyme levels in the absence of any hepatotoxicity or immune reactions.

 

Clinical need:  Currently, most of the therapeutic options are diet-mediated to reduce the amount of glyoxylate intake and maximize the intake of vitamin B6.  The most effective treatment for PH1 is pre-emptive liver transplantation, alone or liver combined with kidney transplantation in ESRF.  There is therefore a clear need for alternative or new treatments options.

 

Feasibility :

 

Gene : the AGXT gene maps onto chromosome 2q36-q37, has a 10 kb coding sequence and contains 11 exons generating a 392-residue protein.

 

Vector optimization: Eight constructs different containing codon optimized AGXT genes have already been generated in-house and are currently being characterized.

 

Animal models: Small animal models already exist and have been used for pre-clinical proof of concept studies.

 

Biomarkers: Measurements of oxalate are part of routine clinical practice for the disease setting and

 



 

monitoring of kidney changes can also be done using standard techniques.

 

After a phase of further vector optimization it is our aim to develop this product for a first Phase I clinical study by mid 2016. Further development timelines have not been defined.

 

Development overview to IMPD:

 

 

The GLP safety and toxicology studies will provide the first milestone (Go/No Go) for the project.

 

Safety Assessment: At this stage is not possible to make any inferences in relation to potential safety concerns.

 

Central Nervous System Programs

 

A)             Development Programs

 

1.               AMT-110 for Sanfilippo B

 

Disease Background

 

Sanfilippo syndrome, or Mucopolysaccharidosis type III (MPSIII), is a rare lysosomal storage disorder (LSD) that occurs when enzymes needed to break down the heparan sulfate sugar chain are missing or are defective. Sanfilippo B is one of the four types of MPSIII that results in serious brain degeneration in children, and is generally lethal. The deficient enzyme responsible for the disease is alpha-N-acetylglucosaminidase (NaGlu). The clinical manifestations are mainly neurological, with early symptoms observed during the first five years of age, leading to a progressive deterioration of cognitive abilities. Affected children require specific care after age seven and progressively develop profound mental retardation with reduced somatic manifestations. Death frequently occurs at the median age of 15. No treatment is currently available.

 

Birth prevalences of 0.28—4.1 per 100, 000 have been reported (Valstar et al., 2008). More recently, He´ron et al. (2010) estimated the mean annual incidence for Sanfilippo B in France at 0.15 per 100,000 births.

 

Overview of AMT-110

 

The goal of our AMT-110 program is to provide a gene therapy for Sanfilippo B syndrome through the introduction of a functional NaGlu gene into the patients’ brain cells.

 



 

This project is being pursued together with the Pasteur Institute (Paris) whereby uniQure is responsible for developing the manufacturing process and producing clinical grade material and the Pasteur Institute for conducting the clinical trials.

 

Preclinical Development

 

·                   Product Profile

 

AMT-110 is designed to be delivered via intracranial administration.

AMT-110 or rAAV5-hNaGlu, is a recombinant adeno-associated vector of serotype 5, consisting of:

 

·                   Inverted terminal regions or ITRs of the adeno-associated serotype 2

·                   A human α-N-acetylglucosaminidase, or hNaGlu, gene the therapeutic gene

·                   The mouse phosphoglycerate kinase-1 promoter (muPGK)

 

·                   Pre-clinical Proof of Concept

 

Preclinical PoC studies were conducted in mouse and dog disease models at the Pasteur Institute. These studies showed that mice with MSPIIIB a single AAV5-NaGlu intracranial injection resulted in reversion of storage lesions throughout the brain and prevented loss of Purkinje cells. Furthermore, it improved animal behavior and corrected pathological featured of the disease including, neuro-inflammation, axonal transport, synaptic vesicle content and the autophagy defect.

 

In MSPIIIB dogs treatment (four simultaneous injections) with AAV5-hNaGlu was well tolerated, vector particles were broadly distributed and the therapeutic enzyme was delivered to the entire brain leading to improvement in MPSIIIB-induced histological lesions and normalization of glycosaminoglycan and ganglioside levels. These studies also confirmed that the combination of gene therapy with efficient immunosuppression was required for treatment efficacy.

 

·                   Non-clinical safety & toxicology studies

 

The following table presents a summary of the AMT-10 non-clinical safety and toxicology studies that have been conducted to support the clinical development program.

 

Parameter to be
assessed

 

Study performed

 

Results

Pharmacokinetics

 

1) GLP biodistribution in Sprague-Dawley rats with or without immunosuppression (180 days)

 

 

 

2) GLP biodistribution in Dogs

 

In in Sprague-Dawley rats:

 

·                   Constant high number of genome copies present in the brain with no vector in the blood at 3 months.

·                   No difference between immunosuppressed and non-

 



 

 

 

with immunosuppression (90 days)

 

                        immuno-suppressed animals. In dogs

·                   Organs negative for vector DNA (< LOQ) were heart, lung, kidney and testis whereas positive organs (> LOQ) included spleen, liver and cervical spinal cord.

 

Toxicity

 

 GLP toxicity study in Sprague-Dawley rats (180 days)

 

(French Authorities endorsed the use of a single species for safety and toxicology studies)

 

 

 

·                   No specific toxicity

·                   Only injection traces in the brain at some injection sites

·                   No effect on body weight, growth, food consumption, behaviour

·                   No decedents, no inflammation

·                   No histopathological findings

·                   NOAEL: 3.4 x 10 11 /animal

 

Persistence in semen and risk of germline transmission

 

 

 

Based on pilot dog preliminary data where no vector DNA was found in the gonads, French Authorities endorsed the proposal that no germline transmission studies were necessary.

 

Summary of AMT-110 Preclinical Development Program

 

·                   In animal models of Sanfilippo B, treatment with AAV5-hNaGlu ameliorated pathophysiological signs and symptoms of the disease.

 

·                   AMT-110 administered into the striatum of non-immunosuppressed rats and immunosuppressed rats and dogs produced long lasting presence of vector DNA in the brain and caused no mortality and no signs of toxicity.

 

Clinical Development Program

 

The key regulatory and clinical development milestones for AMT-110 include the following ,

 



 

·

 

1 st  Scientific Advice with French Regulatory Authorities

 

March 2012

 

 

 

 

 

·

 

2 nd  Scientific Advice with French Regulatory Authorities

 

March 2012

 

 

 

 

 

·

 

IMPD Submission

 

April 2013

 

 

 

 

 

·

 

IMPD Approval

 

3Q13

 

 

 

 

 

·

 

Phase I start

 

October 2013

 

Expected Milestones

 

·

 

Phase II/III start

 

2016

 

 

 

 

 

·

 

Registration

 

2018

 

The Phase I/II study is a single center, open label, prospective, interventional, single dose of AAV5-hFIXco (AMT-060) trial in children with Sanfilippo type B syndrome. Administration will be performed into the brain parenchyma and cerebellum at 8 image-guided tracks, with 2 deposits per tracks, in a single neurosurgical session. Each patient will receive 960 μL of vector suspension as 16 simultaneous vector deposit each containing 2.4 10 11  gc (4 10 12  gc in total). Patients will receive immunosuppression starting 10 days prior to treatment.

 

The primary objective of the study is to evaluate the clinical, radiological and biological safety of the treatment. The secondary objective is to collect samples and data to define exploratory tests that could become evaluation criteria for further clinical efficacy studies (Brain MRI; neurological tests and biological markers).

 

The study will be conducted at the Bicêtre Hospital which is part of the University Hospitals of South Paris and is expected to enroll a total of 4 children during an 8 to 12 months inclusion period. The duration of follow-up for each patient is one year. The first patient was dosed in October 2013.

 

·                   Future Clinical Development

 

Licensee plans to complete the Phase I and start a Phase II/III trial in multiple sites worldwide.  Following initiation of this trial one of the options on how to proceed would be applying for approval for compassionate use to treat on a named patient basis. This can be well justified based on the size of the indication and lethality of the condition.

 

Summary of AMT-110 Clinical Development Program

 

·                   The IMPD was submitted in April 2013

 

·                   Phase I was started in October 2013

 

1.2

 



 

2.               AMT-090 for Parkinson’s Disease

 

Disease Background

 

Parkinson’s Disease (PD) is a progressive neurodegenerative disorder that affects motor skills, speech, and other neurological functions. As the condition progresses, every action becomes increasingly difficult, and eventually impossible. The symptoms are caused by degeneration and death of nerve cells in the substantia nigra, a part of the brain that produces dopamine needed to specifically control body movements.  Dopamine is a chemical that sends messages in the brain to coordinate and control muscular action and movements. There is currently no cure for Parkinson’s Disease, but medications or surgery can provide symptomatic relief, the efficacy of which declines over time and which create significant side effects and co-morbidities, such as depression and dyskinesias. The most widely used treatment is L-dopa in various forms, which is converted to dopamine in the central nervous system. Current symptomatic treatments for Parkinson’s Disease represent a multi-billion dollar market.

 

While medications can temporarily alleviate the symptoms of PD, they do not influence the degenerative process.  Progressive loss of nigral dopaminergic (DA) neurons (the pathological hallmark of PD) results in progressive neurologic dysfunction and death.  Glial cell line-derived neurotrophic factor (GDNF) was first identified based on its ability to promote the survival of embryonic DA neurons in vitro, and research has demonstrated beneficial effects of GDNF in animal models of PD.  Recent evidence indicates that gene transfer via direct delivery of viral vectors may represent a superior approach for the treatment of PD with GDNF.

 

Based on the overwhelming preclinical data of GDNF protective effects on DA neurons, a series of preclinical and clinical studies conducted by third parties have consistently indicated that the infusion of GDNF protein into the brain is effective in Parkinson’s Disease. Three clinical trials were performed with direct infusion into the putamen.  Two of the studies (Bristol, UK and University of Kentucky) reported favorable clinical response (Gill et al., 2003, Patel et al., 2005, Slevin et al., 2005), and one sponsored by Amgen in the US was abandoned due to apparent lack of efficacy and the appearance of neutralizing antibodies to GDNF in some patients (Lang et al., 2006a). The outcome of these GDNF protein trials still remains controversial (Barker, 2006, Chebrolu et al., 2006, Lang et al., 2006a, Lang et al., 2006b, Penn et al., 2006, Slevin et al., 2006), but the consensus in the scientific community seems to be that the cannula used in the Amgen trial was not optimal, leading to a leakage of the protein into cerebrospinal fluid (CSF).  Results from these early clinical trials with GDNF protein underscore the need for a clinical approach in which appropriate levels of GDNF are delivered accurately to the intended sites in the brain where the DA neurons and their terminals reside.  Stereotactic parenchymal convection-enhanced delivery of viral vectors carrying the GDNF gene is more likely to achieve precise delivery.

 

PD is a progressive neurodegenerative disease that advances inexorably over a period of 10 to 30 years to disability and death.  Medications, generally those aimed at ameliorating the known striatal dopamine deficiency, can provide substantial benefits for the cardinal symptoms of PD, namely resting tremor, rigidity, bradykinesia and postural instability.  Unfortunately, the clinical response wanes over time and a variety of medication-related complications emerge including motor fluctuations, dyskinesias, short duration responses, and psychosis.  Disease progression continues since dopamine replacement and other medical therapies have no impact on the underlying neurodegenerative process.  Stereotactic deep brain stimulation has emerged as a rational treatment option, but this surgical approach is also symptomatic only and may be associated with serious adverse effects like stroke, hemorrhage, or infection, and hardware-related complications.

 

Overview of AMT-090 Program

 

Licensee’s AMT-090 program seeks to introduce the gene encoding the GDNF protein to provide a consistent supply of GDNF to the relevant areas of the brain. Our goal is to inject our AAV2 vector carrying the gene for GDNF into the brain to stop the progression of the disease and possibly measurable clinical and neuroimaging improvement. One of the key elements here is the MRI-guided convection enhanced delivery, which ensures for proper targeting of the vector.

 



 

Preclinical Development

 

Initial preclinical research was conducted in partnership with the University of Lund, Sweden, which established proof of concept in rodents. Moreover, the University of California San Francisco (UCSF) has conducted many studies consistently demonstrating a therapeutic effect of GDNF in rodents and primates. Key findings include:

 

·                   successful gene transfer with AAV2 in the Putamen of rats

 

·                   MRI-guided CED delivery of the AAV2 vector to the putamen in non-human primates resulted in GDNF expression in the putamen but also in the substantia nigra

 

·                   in a rat lesion model, AAV-GDNF delivery was able to protect neurons from degeneration

 

·                   in a primate lesion model, AAV-GDNF delivery was able to protect neurons from degeneration

 

·                   no toxicity was observed at any dose levels

 

·                   AAV-GDNF was therapeutic in rodent and primate models

 

Development Program

 

·                   Phase I Clinical Trial

 

uniQure has entered into an agreement with UCSF and the National Institute of Neurological Diseases and Stroke. Under this agreement, UCSF commenced a Phase I trial of an AAV2 glial cell line-derived neurotrophic factor (GDNF) treatment for Parkinson’s Disease in May 2013. This trial is being funded by the National Institutes of Health. Licensee has an exclusive right from UCSF to obtain all data related to the program.

 

The trial includes 24 patients afflicted with advanced Parkinson’s Disease (Hoehn and Yahr Stage III or IV off medication) with a Unified PD Rating Scale (UPDRS) (Fahn et al., 1987) total motor score > 30 in the defined off state and a serum anti-AAV2 total antibody titer <1000.

 

The study will entail a Phase 1 single-center, open-label, dose escalation, safety and tolerability study of adeno-associated virus, serotype 2 vector (AAV2) containing human GDNF complementary DNA bilaterally delivered by MRI-guided convection-enhanced delivery (CED) to the putamen (450 µl per hemisphere) of the.  Four escalating dose levels will be evaluated in the following dose cohorts (6 patients per cohort):  Cohort 1 = 9 x 10 10  vg, Cohort 2 = 3 x 10 11  vg, Cohort 3 = 9 x 10 11  vg and Cohort 4 = 3 x 10 12  vg.

 

The trial’s primary objectives are to assess the safety and tolerability of 4 different dose levels of AAV2-GDNF. The secondary objectives of the trial are to obtain preliminary data regarding the potential for clinical responses of the 4 dose levels tested by assessing the magnitude and variability of any treatment effects (via clinical, laboratory and neuroimaging studies).

 

·                   Future Clinical Development

 

Licensee intends to transition this program to our vector and manufacturing platforms and continue the clinical development program. Bridging of the vectors shall include at least testing in Parkinsonian primates and is anticipated to start one year after the first injections in patients are performed. This will then be followed by a multicenter randomized (delayed) start, blinded, sham-controlled Phase 2 efficacy study of this experimental therapy with our vector.  However, prior to finalizing the design of such a trial, Licensee propose sto conduct a preliminary clinical study that should provide critical information for translating the laboratory research to investigations

 



 

involving human subjects and critical data for finalizing the ultimate efficacy trial protocol.  The preliminary study will also allow us to develop the organizational and logistical processes that will be needed for the anticipated multicenter efficacy trial.

 

Summary of AMT-090 Clinical Development Program

 

·                   uniQure has licensed the GDNF gene from Amgen

 

·                   Pre-clinical PoC studies have been conducted in rodents and non-human primates in partnership with the University of Lund (Sweden) and UCF

 

·                   A Phase I human trial in Parkinson’s disease with AAV2 delivering GDNF has been initiated through a partnership with UCSF

 

·                   Initiation of first Phase I clinical trial or foreign equivalent — May 2013

 

Expected milestones

 

·                   Initiation of first Phase I clinical trial or foreign equivalent — 2nd quarter 2013

 

·                   Initiation of first Phase II clinical trial or foreign equivalent — 2nd quarter 2017

 

·                   Initiation of first Phase III clinical trial or foreign equivalent — 4th quarter 2020

 

·                   Submission to Regulatory Authority of First Marketing Approval or foreign equivalent — 2024

 



 

B) Active Research Projects

 

1.               Huntington’s Disease

 

Disease background : Huntington’s Disease (HD) is a neurodegenerative genetic disorder that affects motor control and leads to cognitive decline and dementia. It typically becomes noticeable in middle age, but can begin at any age from infancy to old age. HD has a prevalence of around 1 affected individual in 100,000.

 

The mutated form of the protein huntingtin causes cellular dysfunction and death in a number of CNS sites but is most noticeable in the striatum and cortex. The mutation is caused by CAG repeats in the DNA of patients. The earliest features of HD are involuntary movements and irritability and a loss of executive function. This progresses over time and in the more advanced stages, the patient is demented and bed-bound. The disease is currently incurable with patients dying about 20-25 years after it begins.

 

Clinical need : The clinical need for these patients is high as there is no cure for the disease.

 

Feasibility

 

As the CAG repeats in the Huntingtin gene are the cause of the disease, downregulation of the expression of the CAG repeats is an option. Also rescuing the neurons from degeneration using GDNF is an option. Both options are currently under investigation. Replacing the gene is not an option as this is far too large to fit into an AAV vector.

 

Several transgenic mice models exist. Severity and time of onset are based on the number of CAG repeats in the model. Mostly used are the R6/1 and R6/2 transgenic models.

 

Preclinical work: Proof of concept using GDNF has been established in one laboratory. Licensee iscurrently trying to establish this with our own vector in the laboratory of Roger Barker.

 

Proof of concept with siRNA has been established in mice models and Licensee is in the process of implementing this into our studies.

 

Development overview to IMPD:

 

 

The proof of concept studies ( in vivo/ in vitro work) will provide the first milestone (Go/No Go) for the project.

 

With regards to the siRNA approach to HD, vector generation & optimization will require an additional 9 months prior to any other activity.  Then a similar development path to what is shown above will need to be followed.

 

It is Licensee’s aim upon a successful PoC to develop this product further to a Phase I clinical investigation which should start mid 2016.

 

Collaborators: Licensee is working together with Roger Barker (Cambridge University) on the use of GDNF to rescue neurons in Huntington models, based on a EUREKA grant. Licensee is also working together with Nicole

 



 

Deglon (Lausanne University), Anna Skorupska (Lublin University) and Sebastian Kuegler (Gottingen University) in a Eurostars grant setting. Competition comes from siRNA companies.

 

Safety concerns: Potential safety concerns could be the complete downregulation of the Huntingtin gene, even though not fully supported by the Eurostars team. The use of GDNF could lead to side effects, such as weight loss.

 

IP: For GDNF, Licensee has a license from Amgen. For the siRNA work Licensee has a non-exclusive license from Benitec.

 

2.               Multiple System Atrophy

 

Disease Background: Multiple System Atrophy (MSA) is a sporadic neurodegenerative disease that is characterized by the presence of glial inclusion bodies, which stain positive for a synuclein. The clinical picture is that of parkinsonism, autonomic failure, cerebellar ataxia and pyramidal signs in differing combinations. Approximately 80% of patients present with predominantly parkinsonian features (MSA-P) manifesting in rapidly deteriorating akinesia, rigidity, postural instability and high pitched dysarthria.  Most such patients do not exhibit the classic resting tremor of Parkinson’s disease and virtually all develop frank dysautonomia in the course of the illness.  The cause of the disease is not known.

 

Clinical need: Although a minority of patients may achieve modest benefit from dopaminergic therapy, there is no satisfactory treatment for the parkinsonian disabilities of MSA-P. Additionally, deep brain stimulation of the subthalamic nucleus has been of little or no value. Within 5 years of disease onset patients die so the clinical need is high for these patients.

 

Feasibility:

 

MSA is not a single monogenic disease, but may be treated with a single neuroprotective protein. In this case, this could be GDNF. Some transgenic animal models exist, all overexpressing the alpha-synuclein protein. The rationale to use GDNF (besides its general neuroprotective effect on neurons) is that both in patients and the transgenic mouse model, GDNF expression is downregulated. Introduction of an elevated level of GDNF may serve as the treatment. Read out parameters for the disease progression are all related to those of Parkinson’s Disease. PoC has not yet been established, but is under investigation in the mouse model.

 

Development overview to IMPD:

 

 

The proof of concept studies ( in vivo/ in vitro work) will provide the first milestone (Go/No Go) for the project.

 

It is our aim upon a successful PoC to develop this product further to a Phase I clinical investigation which should start mid 2016.

 

Collaborators: Licensee is working together with Erwan Bezard (University of Bordeaux) and Olivier Rascol (University of Toulouse) who are together running the French reference center for MSA.

 

Safety Assessment: The use of GDNF could lead to side effects, such as weight loss. The exact mechanism through

 



 

which the treatments would have its effect is not clear yet.

 

3.               Hearing loss

 

Disease background : Hearing loss is a serious clinical problem. Underlying mechanisms for the loss of neurons in the cochlea can vary from ischemia, mechanical stress to toxic insults. The actual numbers of patients is not easy to define, but it could be rather large. When age-related hearing loss is also taken into account, this is no longer an orphan indication.

 

Clinical need : Patients with hearing loss could be helped with cochlear implants. However, progressive neurodegeneration is not stopped by that. There is high clinical need as there is no cure for the disease.

 

Feasibility:

 

Neuron function and survival is dependent on a delicate balance of neurotrophins. Following trauma or toxic insult to neurons, they may slowly die. To reverse this state of degeneration, it could be beneficial to supply the neurons with a neurotrophin such as GDNF. This neurotrophin has been shown to be able to rescue neurons from degeneration in several models, including those of the substantia nigra and for instance motorneurons in the spinal cord after trauma.

 

Animal models are available and include for instance use of Kanamycin in cats, mice or guinea pigs. Also chemotherapeutic agents from the class of statins are used.

 

Preclinical work: Proof of concept using recombinant brain-derived neurotrophic factor (BDNF) and/or GDNF has been established. Licensee is currently trying to establish this with our own vector in the laboratory of Patricia Leake.

 

Cochlea of mice can be transduced to express a recombinant transgene.

 

Development overview to IMPD:

 

 

The proof of concept studies ( in vivo/ in vitro work) will provide the first milestone (Go/No Go) for the project.

 

This new project has just been initiated upon a successful PoC it is our aim to develop this product further to a Phase I clinical trial, which should start by the end 2016.

 

Collaborators: Licensee is working together with Patricia Leake (University College of San Francisco) on the use of GDNF to rescue neurons in mouse and cat models. She is the investigator who developed the cochlear implant. This could also be included in the experimental plan.

 

Safety concerns: The use of GDNF could lead to side effects. Weight loss is not expected, but as the GDNF also has a neurotrophic effect, nerve fibers could sprout in an aberrant way possibly leading to incorrect connections.

 



 

IP: For GDNF, Licensee has a license from Amgen; the program as a whole is under investigation.

 

Exploratory Research Projects

 

The projects listed under this category in Table 1 above are not in active research yet, but are likely targets for our platform technology and are being assessed on feasibility before starting active bench work.

 


Exhibit 10.2

 

PUBLIC HEALTH SERVICE

 

PATENT LICENSE AGREEMENT - EXCLUSIVE and NON-EXCLUSIVE

 

COVER PAGE

 

For PHS internal use only:

 

License Number:
L-116-2011/0

 

License Application Number: A-063-2009

 

Serial Number(s) of Licensed Patent(s) or Patent Application(s):

 

1. U.S. Patent 6,984,517, issued January 10, 2006, entitled “AAV5 Vector and Uses Thereof; U.S. Patent 7,479,554, issued January 20, 2009, entitled “AAV5 Nucleic Acids” as well as all issued and pending foreign counterparts [ HHS Ref. No. E-127-1998/0];

 

2. U. S. Patent 6,855,314, issued February 15, 2005, entitled “AAV5 Vector for Transducing Brain Cells and Lung Cells” [ HHS Ref. No. E-072-2000/0]

 

Licensee: Amsterdam Molecular Therapeutics (AMT) B.V.

 

Cooperative Research and Development Agreement (CRADA) Number (if a subject invention): N/A

 

Additional Remarks: This Patent License Agreement will replace PHS license L-119-2007/0 and any amendments thereto.

 

Public Benefit(s): Commercialization of this technology will benefit the public health by providing AAV5 based gene therapies to treat diseases originated from the brain and liver.

 

This Patent License Agreement, hereinafter referred to as the “ Agreement ”, consists of this Cover Page, an attached Agreement , a Signature Page, Appendix A (List of Patent(s) or Patent Application(s)), Appendix B (Fields of Use and Territory), Appendix C (Royalties), Appendix D (Benchmarks and Performance), Appendix E (Commercial Development Plan), Appendix F (Example Royalty Report), and Appendix G (Royalty Payment Options).  The Parties to this Agreement are:

 

1)                                      The National Institutes of Health (“ NIH ”) or the Food and Drug Administration (“ FDA ”), hereinafter singly or collectively referred to as “ PHS ”, agencies of the United States Public Health Service within the Department of Health and Human Services (“ HHS ”); and

 

2)                                      The person, corporation, or institution identified above or on the Signature Page, having offices at the address indicated on the Signature Page, hereinafter referred to as “ Licensee ”.

 



 

PHS PATENT LICENSE AGREEMENT - EXCLUSIVE and NON-EXCLUSIVE

 

PHS and Licensee agree as follows:

 

1.                                       BACKGROUND

 

1.1                                In the course of conducting biomedical and behavioral research, PHS investigators made inventions that may have commercial applicability.

 

1.2                                By assignment of rights from PHS employees and other inventors, HHS , on behalf of the Government , owns intellectual property rights claimed in any United States or foreign patent applications or patents corresponding to the assigned inventions.  HHS also owns any tangible embodiments of these inventions actually reduced to practice by PHS .

 

1.3                                The Secretary of HHS has delegated to PHS the authority to enter into this Agreement for the licensing of rights to these inventions.

 

1.4                                PHS desires to transfer these inventions to the private sector through commercialization licenses to facilitate the commercial development of products and processes for public use and benefit.

 

1.5                                Licensee desires to acquire commercialization rights to certain of these inventions in order to develop processes, methods, or marketable products for public use and benefit.

 

2.                                       DEFINITIONS

 

2.1                                Affiliate(s) ” means a corporation or other business entity, which directly or indirectly is controlled by or controls, or is under common control with Licensee .  For this purpose, the term “control” shall mean ownership of more than fifty percent (50%) of the voting stock or other ownership interest of the corporation or other business entity, or the power to elect or appoint more than fifty percent (50%) of the members of the governing body of the corporation or other business entity.

 

2.2                                Benchmarks ” mean the performance milestones that are set forth in Appendix D.

 

2.3                                Commercial Development Plan ” means the written commercialization plan attached as Appendix E.

 

2.4                                Exempt Collaborator ” means a not-for-profit organization or academic institution that has entered into a formal collaboration and / or supply agreement with Licensee to conduct pre-clinical development and solely sponsor clinical trials of Licensed Product , excluding Supplied Materials , to treat an Ultra-Orphan Indication ; in which Licensee may acquire clinical development and data for regulatory approval and sale of a Licensed Product .

 

2.5                                First Commercial Sale ” means the initial transfer by or on behalf of Licensee or its sublicensees of Licensed Products or the initial practice of a Licensed Process by or on behalf of Licensee or its sublicensees in exchange for cash or some equivalent to which value can be assigned for the purpose of determining Net Sales .

 

2.6                                Government ” means the Government of the United States of America.

 

2.7                                Licensed Fields of Use ” means the fields of use a) and b) as identified in Appendix B.

 

2.8                                Licensed Patent Rights ” shall mean:

 



 

(a)                                  Patent applications (including provisional patent applications and PCT patent applications) or patents listed in Appendix A, all divisions and continuations of these applications, all patents issuing from these applications, divisions, and continuations, and any reissues, reexaminations, and extensions of these patents;

 

(b)                                  to the extent that the following contain one or more claims directed to the invention or inventions disclosed in 2.8(a):

 

(i)                                      continuations-in-part of 2.8(a);

 

(ii)                                   all divisions and continuations of these continuations-in-part;

 

(iii)                                all patents issuing from these continuations-in-part, divisions, and continuations;

 

(iv)                               priority patent application(s) of 2.8(a); and

 

(v)                                  any reissues, reexaminations, and extensions of these patents;

 

(c)                                   to the extent that the following contain one or more claims directed to the invention or inventions disclosed in 2.8(a): all counterpart foreign and U.S. patent applications and patents to 2.8(a) and 2.8(b), including those listed in Appendix A; and

 

(d)                                  Licensed Patent Rights shall not include 2.8(b) or 2.8(c) to the extent that they contain one or more claims directed to new matter which is not the subject matter disclosed in 2.8(a).

 

2.9                                Licensed Processes ” means processes which, in the course of being practiced, would be within the scope of one or more claims of the Licensed Patent Rights that have not been held unpatentable, invalid or unenforceable by an unappealed or unappealable judgment of a court of competent jurisdiction.

 

2.10                         Licensed Products ” means tangible materials which, in the course of manufacture, use, sale, or importation, would be within the scope of one or more claims of the Licensed Patent Rights that have not been held unpatentable, invalid or unenforceable by an unappealed or unappealable judgment of a court of competent jurisdiction.

 

2.11                         Licensed Territory ” means the geographical area identified in Appendix B.

 

2.12                         Marketing Approval ” means any and all approvals (including price and reimbursement approvals, if required), licenses, registrations, or authorizations of regulatory authorities in any country that are necessary for the manufacture, use, storage, import, transport and/or sale of a Licensed Product in the Licensed Fields of Use in such country.

 

2.13                         Net Sales ” means the total gross receipts for sales of Licensed Products or practice of Licensed Processes by or on behalf of Licensee or its sublicensees, and from leasing, renting, or otherwise making Licensed Products available to others without sale or other dispositions, whether invoiced or not, less returns and allowances, packing costs, insurance costs, freight out, taxes or excise duties imposed on the transaction (if separately invoiced), and wholesaler and cash discounts in amounts customary in the trade to the extent actually granted.  No deductions shall be made for commissions paid to individuals, whether they are with independent sales agencies or regularly employed by Licensee , or sublicensees, and on its payroll, or for the cost of collections.

 



 

2.14                         Orphan Indication ” means a disease that affects less than two hundred thousand (200,000) people in the United States as defined by the Food and Drug Administration or five (5) in ten thousand (10,000) people in the European Union as defined by the European Medicines Agency.

 

2.15                         Practical Application ” means to manufacture in the case of a composition or product, to practice in the case of a process or method, or to operate in the case of a machine or system; and in each case, under these conditions as to establish that the invention is being utilized and that its benefits are to the extent permitted by law or Government regulations available to the public on reasonable terms.

 

2.16                         Research License ” means a nontransferable, nonexclusive license to make and to use Licensed Products or Licensed Processes as defined by the Licensed Patent Rights for purposes of research and not for purposes of commercial manufacture or distribution or in lieu of purchase.

 

2.17                         Supplied Materials ” means (a) A helper plasmid for AAV5 5RepCap 5RepCapB containing the p5 promoter, the AAV5 Rep and AAV5 Cap genes and an SV40ori adjacent to the polyadenylation signal at the 3’ end or equivalent; (b) A vector plasmid for AAV5, pAAV5LacZ/pAAV5RnLacZ expressing nucleus localized beta-galactosidase contains the LacZ gene under control of Rous Sarcoma Virus (RSV) promoter between the AAV5 ITRs or equivalent both of which are described in Chiorini et at Virol;. 73(2):1309-19 (Feb. 1999), including any progeny, subclones, or unmodified derivatives thereof where “unmodified derivatives” is defined as set forth in the Uniform Biological Material Transfer Agreement as published in the Federal Register at 60(45): 12771-75 (March 8, 1995); and (c) Plasmid maps corresponding to items (a) and (b) as set forth in the Paragraph.  Further, these Supplied Materials were supplied by PHS to Licensee under a Material Transfer Agreement.

 

2.18                         Third Party Applicant ” shall mean any non- Licensee applicant from whom PHS receives a license application for Licensed Patent Rights in an indication for which proposed commercial development is not addressed in Licensee ’s then current Commercial Development Plan outlined in Appendix E of this Agreement .

 

2.19                         Ultra-Orphan Indication ” means a disease that affects less than one (1) in Fifty Thousand (50,000) people in the United States or the European Union.

 

3.                                       GRANT OF RIGHTS

 

3.1                                PHS hereby grants and Licensee accepts, subject to the terms and conditions of this Agreement , an exclusive license and non-exclusive license, as specified in Appendix B, under the Licensed Patent Rights in the Licensed Territory to make and have made, to use and have used, to sell and have sold, to offer to sell, and to import any Licensed Products in the Licensed Fields of Use and to practice and have practiced any Licensed Processes in the Licensed Fields of Use .

 

3.2                                This Agreement confers no license or rights by implication, estoppel, or otherwise under any patent applications or patents of PHS other than the Licensed Patent Rights regardless of whether these patents are dominant or subordinate to the Licensed Patent Rights .

 

4.                                       SUBLICENSING

 

4.1                                Upon written approval, which shall include prior review of any sublicense agreement by PHS and which shall not be unreasonably withheld, Licensee may enter into sublicensing agreements under the Licensed Patent Rights .

 

4.2                                Licensee agrees that any sublicenses granted by it shall provide that the obligations to PHS of Paragraphs 5.1-5.4, 8.1, 10.1, 10.2, 12.5, and 13.8-13.10 of this Agreement shall be binding upon

 



 

the sublicensee as if it were a party to this Agreement Licensee further agrees to attach copies of these Paragraphs to all sublicense agreements.

 

4.3                                Any sublicenses granted by Licensee shall provide for the termination of the sublicense, or the conversion to a license directly between the sublicensees and PHS , at the option of the sublicensee, upon termination of this Agreement under Article 13.  This conversion is subject to PHS approval (not to be unreasonably withheld) and contingent upon acceptance by the sublicensee of the remaining provisions of this Agreement .

 

4.4                                Licensee agrees to forward to PHS a complete copy of each fully executed sublicense agreement postmarked within thirty (30) days of the execution of the agreement.  To the extent permitted by law, PHS agrees to maintain each sublicense agreement in confidence.

 

5.                                       STATUTORY AND PHS REQUIREMENTS AND RESERVED GOVERNMENT RIGHTS

 

5.1                                (a)                                  PHS reserves on behalf of the Government an irrevocable, nonexclusive, nontransferable, royalty-free license for the practice of all inventions licensed under the Licensed Patent Rights throughout the world by or on behalf of the Government and on behalf of any foreign government or international organization pursuant to any existing or future treaty or agreement to which the Government is a signatory.  Prior to the First Commercial Sale , Licensee agrees to provide PHS with reasonable quantities of Licensed Products or materials made through the Licensed Processes for PHS research use; and

 

(b)                                  In the event that the Licensed Patent Rights are Subject Inventions made under a Cooperative Research and Development Agreement (“ CRADA ”), Licensee grants to the Government , pursuant to 15 U.S.C. §3710a(b)(l)(A), a nonexclusive, nontransferable, irrevocable, paid-up license to practice Licensed Patent Rights or have Licensed Patent Rights practiced throughout the world by or on behalf of the Government .  In the exercise of this license, the Government shall not publicly disclose trade secrets or commercial or financial information that is privileged or confidential within the meaning of 5 U.S.C. §552(b)(4) or which would be considered as such if it had been obtained from a non-Federal party.  Prior to the First Commercial Sale , Licensee agrees to provide PHS reasonable quantities of Licensed Products or materials made through the Licensed Processes for PHS research use.

 

5.2                                Licensee agrees that products used or sold in the United States embodying Licensed Products or produced through use of Licensed Processes shall be manufactured substantially in the United States, unless a written waiver is obtained in advance from PHS .

 

5.3                                Licensee acknowledges that PHS may enter into future CRADAs under the Federal Technology Transfer Act of 1986 that relate to the subject matter of this Agreement Licensee agrees not to unreasonably deny requests for a Research License from future collaborators with PHS when acquiring these rights is necessary in order to make a CRADA project feasible.  Licensee may request an opportunity to join as a party to the proposed CRADA .

 

5.4                                (a)                                  In addition to the reserved license of Paragraph 5.1, PHS reserves the right to grant Research License s directly or to require Licensee to grant Research License s on reasonable terms.  The purpose of these Research License s is to encourage basic research, whether conducted at an academic or corporate facility.  In order to safeguard the Licensed Patent Rights , however, PHS shall consult with Licensee before granting to commercial entities a Research License or providing to them research samples of materials made through the Licensed Processes ; and

 



 

(b)                                  In exceptional circumstances, and in the event that Licensed Patent Rights are Subject Inventions made under a CRADA , the Government , pursuant to 15 U.S.C. §3710a(b)(l)(B), retains the right to require the Licensee to grant to a responsible applicant a nonexclusive, partially exclusive, or exclusive sublicense to use the Licensed Patent Rights in the Licensed Field of Use on terms that are reasonable under the circumstances, or if Licensee fails to grant this license, the Government retains the right to grant the license itself.  The exercise of these rights by the Government shall only be in exceptional circumstances and only if the Government determines:

 

(i)                                      the action is necessary to meet health or safety needs that are not reasonably satisfied by Licensee ;

 

(ii)                                   the action is necessary to meet requirements for public use specified by Federal regulations, and these requirements are not reasonably satisfied by the Licensee ; or

 

(iii)                                the Licensee has failed to comply with an agreement containing provisions described in 15 U.S.C. §3710a(c)(4)(B); and

 

(c)                                   The determination made by the Government under this Paragraph 5.4 is subject to administrative appeal and judicial review under 35 U.S.C. §203(b).

 

6.                                       ROYALTIES AND REIMBURSEMENT

 

6.1                                Licensee agrees to pay PHS a noncreditable, nonrefundable license issue royalty as set forth in Appendix C.

 

6.2                                Licensee agrees to pay PHS a nonrefundable minimum annual royalty as set forth in Appendix C.

 

6.3                                Unless otherwise exempted in Paragraphs 6.13-6.19, Licensee agrees to pay PHS earned royalties as set forth in Appendix C.

 

6.4                                Unless otherwise exempted in Paragraphs 6.13-6.19, Licensee agrees to pay PHS benchmark royalties as set forth in Appendix C.

 

6.5                                Licensee agrees to pay PHS sublicensing royalties as set forth in Appendix C.

 

6.6                                A patent or patent application licensed under this Agreement shall cease to fall within the Licensed Patent Rights for the purpose of computing earned royalty payments in any given country on the earliest of the dates that:

 

(a)                                  the application has been abandoned and not continued;

 

(b)                                  the patent expires or irrevocably lapses, or

 

(c)                                   the patent has been held to be invalid or unenforceable by an unappealed or unappealable decision of a court of competent jurisdiction or administrative agency.

 

6.7                                No multiple royalties shall be payable because any Licensed Products or Licensed Processes are covered by more than one of the Licensed Patent Rights .  In the event that this Agreement and PHS license L-107-2007/0 as amended from time to time apply to the same product sold by the

 



 

Licensee or its sublicensees then the Licensee shall only pay earned royalties and benchmark royalties under this Agreement .

 

6.8                                On sales of Licensed Products by Licensee to sublicensees or on sales made in other than an arms-length transaction, the value of the Net Sales attributed under this Article 6 to this transaction shall be that which would have been received in an arms-length transaction, based on sales of like quantity and quality products on or about the time of this transaction.

 

6.9                                With regard to unreimbursed expenses associated with the preparation, filing, prosecution, and maintenance of all patent applications and patents included within the Licensed Patent Rights and paid by PHS prior to the effective date of this Agreement Licensee shall pay PHS , as an additional royalty, on or before March 1, 2012, and upon PHS ’ submission of a statement and request for payment to Licensee , an amount equivalent to these unreimbursed expenses previously paid by PHS , the total amount should not exceed two hundred and fifty thousand U.S. dollars ($250,000).  If this Agreement is terminated by Licensee on or before March 1, 2012, Licensee agrees to pay the amount in full within sixty (60) days before termination.

 

6.10                         With regard to unreimbursed expenses associated with the preparation, filing, prosecution, and maintenance of all patent applications and patents included within the Licensed Patent Rights and paid by PHS on or after the effective date of this Agreement PHS , at its sole option, may require Licensee :

 

(a)                                  to pay PHS on an annual basis, within sixty (60) days of PHS ’ submission of a statement and request for payment, a royalty amount equivalent to these unreimbursed expenses paid during the previous calendar year;

 

(b)                                  to pay these unreimbursed expenses directly to the law firm employed by PHS to handle these functions.  However, in this event, PHS and not Licensee shall be the client of the law firm; or

 

(c)                                   in limited circumstances, Licensee may be given the right to assume responsibility for the preparation, filing, prosecution, or maintenance of any patent application or patent included with the Licensed Patent Rights .  In that event, Licensee shall directly pay the attorneys or agents engaged to prepare, file, prosecute, or maintain these patent applications or patents and shall provide PHS with copies of each invoice associated with these services as well as documentation that these invoices have been paid.

 

6.11                         PHS agrees, upon written request, to provide Licensee with summaries of patent prosecution invoices for which PHS has requested payment from the Licensee under Paragraphs 6.9 and 6.10.  Licensee agrees that all information provided by PHS related to patent prosecution costs shall be treated as confidential commercial information and shall not be released to a third party except as required by law or a court of competent jurisdiction.

 

6.12                         Licensee may elect to surrender its rights in any country of the Licensed Territory under any of the Licensed Patent Rights upon sixty (60) days written notice to PHS and owe no payment obligation under Paragraph 6.10 for patent-related expenses paid in that country after ninety (90) days of the effective date of the written notice.

 

6.13                         Exemption for Ultra-Orphan Indication Research

 

(a)                                  Licensee shall be permitted, upon PHS consent, (not to be unreasonably withheld), to manufacture and supply Licensed Product , excluding Supplied Materials , to an Exempt Collaborator for use solely in pre-clinical and clinical development to treat an

 



 

Ultra-Orphan Indication .  Prior to commencement of manufacturing of Licensed Product for an Exempt Collaborator, Licensee shall request permission in writing and must obtain written consent from PHS .  Additional documentation to establish an Exempt Collaborator may be required by PHS .

 

(b)                                  For avoidance of doubt, Licensee shall retain Supplied Materials and shall not release Supplied Materials alone to an Exempt Collaborator .

 

(c)                                   Upon receipt of written consent from PHS for manufacturing of a Licensed Product for an Exempt Collaborator Licensee shall not be obligated to pay Benchmark royalties which would have been payable under Appendix C , Section IV for Benchmarks triggered by clinical trials solely sponsored by the Exempt Collaborator until such time as Licensee exercises its option to acquire the clinical development from the Exempt Collaborator .

 

(d)                                  Upon acquisition of the clinical development from an Exempt Collaborator Licensee shall pay PHS royalties which become payable from that point onwards in accordance with Appendix C , Section IV.  Licensee must inform PHS in writing within thirty (30) days of Licensee ’s decision to acquire or not acquire clinical development from the Exempt Collaborator.

 

(e)                                   For avoidance of doubt, PHS shall consider Licensee ’s sponsorship or co-sponsorship of a clinical trial or regulatory submission for a Licensed Product to treat an Ultra-Orphan Indication as an acquisition of clinical development from an Exempt Collaborator .

 

(f)                                    Earned royalty payments on Net Sales specified in Appendix C , Section III shall not be applicable to Licensed Product manufactured for research and clinical trials conducted by an Exempt Collaborator approved by PHS per Paragraph 6.13.

 

(g)                                   In lieu of earned royalty payments, Licensee shall pay PHS a royalty payment of ten thousand U.S. dollars ($10,000) for each collaboration approved by PHS with an Exempt Collaborator .  Such royalty shall be due within thirty (30) days of the date of PHS written consent per Paragraph 6.13.  In the event that several licenses granted by PHS to the Licensee apply to the same product, only a single payment of $10,000 will be payable per collaboration.

 

7.                                       PATENT FILING, PROSECUTION, AND MAINTENANCE

 

7.1                                Except as otherwise provided in this Article 7, PHS agrees to take responsibility for, but to consult with, the Licensee in the preparation, filing, prosecution, and maintenance of any and all patent applications or patents included in the Licensed Patent Rights and shall furnish copies of relevant patent-related documents to Licensee .

 

7.2                                Upon PHS ’ written request, Licensee shall assume the responsibility for the preparation, filing, prosecution, and maintenance of any and all patent applications or patents included in the Licensed Patent Rights and shall, on an ongoing basis, promptly furnish copies of all patent-related documents to PHS .  In this event, Licensee shall, subject to the prior approval of PHS , select registered patent attorneys or patent agents to provide these services on behalf of Licensee and PHS PHS shall provide appropriate powers of attorney and other documents necessary to undertake this action to the patent attorneys or patent agents providing these services.  Licensee and its attorneys or agents shall consult with PHS in all material aspects of the preparation, filing, prosecution and maintenance of patent applications and patents included within the Licensed

 



 

Patent Rights and shall provide PHS sufficient opportunity to comment on any document that Licensee intends to file or to cause to be filed with the relevant intellectual property or patent office.

 

7.3                                At any time, PHS may provide Licensee with written notice that PHS wishes to assume control of the preparation, filing, prosecution, and maintenance of any and all patent applications or patents included in the Licensed Patent Rights such that the terms of Paragraph 7.1 shall then apply.  If PHS elects to reassume these responsibilities, Licensee agrees to cooperate fully with PHS , its attorneys, and agents in the preparation, filing, prosecution, and maintenance of any and all patent applications or patents included in the Licensed Patent Rights and to provide PHS with complete copies of any and all documents or other materials that PHS deems necessary to undertake such responsibilities.  Licensee shall be responsible for all costs associated with transferring patent prosecution responsibilities to an attorney or agent of PHS ’ choice.

 

7.4                                Each party shall promptly inform the other as to all matters that come to its attention that may materially affect the preparation, filing, prosecution, or maintenance of the Licensed Patent Rights and permit each other to provide comments and suggestions with respect to the preparation, filing, prosecution, and maintenance of Licensed Patent Rights , which comments and suggestions shall be considered by the other party .

 

8.                                       RECORD KEEPING

 

8.1                                Licensee agrees to keep accurate and correct records of Licensed Products made, used, sold, or imported and Licensed Processes practiced under this Agreement appropriate to determine the amount of royalties due PHS .  These records shall be retained for at least five (5) years following a given reporting period and shall be available during normal business hours for inspection, at the expense of PHS , by an accountant selected by PHS for the sole purpose of verifying reports and royalty payments hereunder.  The accountant shall only disclose to PHS information relating to the accuracy of reports and royalty payments made under this Agreement .  Such inspections may be made no more than once each calendar year, with reasonable efforts to minimize disruption of Licensee ’s normal business activities.  Such records for any particular calendar quarter shall be subject to no more than one (1) inspection.  If an inspection shows an underreporting or underpayment in excess of five percent (5%) for any twelve (12) month period, then Licensee shall reimburse PHS for the cost of the inspection at the time Licensee pays the unreported royalties, including any additional royalties as required by Paragraph 9.8.  All royalty payments required under this Paragraph shall be due within sixty (60) days of the date PHS provides Licensee notice of the payment due.

 

9.                                       REPORTS ON PROGRESS, BENCHMARKS, SALES, AND PAYMENTS

 

9.1                                Prior to signing this Agreement , Licensee has provided PHS with the Commercial Development Plan in Appendix E, under which Licensee intends to bring the subject matter of the Licensed Patent Rights to the point of Practical Application .  This Commercial Development Plan is hereby incorporated by reference into this Agreement .  Based on this plan, performance Benchmarks are determined as specified in Appendix D.

 

9.2                                Licensee shall provide written annual reports on its product development progress or efforts to commercialize under the Commercial Development Plan for each of the Licensed Fields of Use within sixty (60) days after December 31 of each calendar year.  These progress reports shall include, but not be limited to: progress on research and development, status of applications for regulatory approvals, manufacturing, sublicensing, marketing, importing, and sales during the preceding calendar year, as well as, plans for the present calendar year.  PHS also encourages these reports to include information on any of Licensee ’s public service activities that relate to the Licensed Patent Rights .  If reported progress differs from that projected in the Commercial

 



 

Development Plan and Benchmarks , Licensee shall explain the reasons for these differences.  In the annual report, Licensee may propose amendments to the Commercial Development Plan , acceptance of which by PHS may not be denied unreasonably.  Licensee agrees to provide any additional information reasonably required by PHS to evaluate Licensee ’s performance under this Agreement Licensee may amend the Benchmarks at any time upon written approval by PHS PHS shall not unreasonably withhold approval of any request of Licensee to extend the time periods of this schedule if the request is supported by a reasonable showing by Licensee of diligence in its performance under the Commercial Development Plan and toward bringing the Licensed Products to the point of Practical Application as defined in 37 C.F.R. §404.3(d).  Licensee shall amend the Commercial Development Plan and Benchmarks at the request of PHS to address any Licensed Fields of Use not specifically addressed in the plan originally submitted.

 

9.3                                Licensee shall report to PHS the dates for achieving Benchmarks specified in Appendix D and the First Commercial Sale in each country in the Licensed Territory within thirty (30) days of such occurrences.

 

9.4                                Licensee shall submit to PHS , within sixty (60) days after each calendar half-year ending June 30 and December 31, a royalty report, as described in the example in Appendix F, setting forth for the preceding half-year period the amount of the Licensed Products sold or Licensed Processes practiced by or on behalf of Licensee in each country within the Licensed Territory , the Net Sales , and the amount of royalty accordingly due.  With each royalty report, Licensee shall submit payment of earned royalties due.  If no earned royalties are due to PHS for any reporting period, the written report shall so state.  The royalty report shall be certified as correct by an authorized officer of Licensee and shall include a detailed listing of all deductions made under Paragraph 2.13 to determine Net Sales made under Article 6 to determine royalties due.

 

9.5                                Licensee agrees to forward semi-annually to PHS a copy of these reports received by Licensee from its sublicensees during the preceding half-year period as shall be pertinent to a royalty accounting to PHS by Licensee for activities under the sublicense.

 

9.6                                Royalties due under Article 6 shall be paid in U.S. dollars and payment options are listed in Appendix G.  For conversion of foreign currency to U.S. dollars, the conversion rate shall be the New York foreign exchange rate quoted in The Wall Street Journal on the day that the payment is due.  Any loss of exchange, value, taxes, or other expenses incurred in the transfer or conversion to U.S. dollars shall be paid entirely by Licensee .  The royalty report required by Paragraph 9.4 shall be mailed to PHS at its address for Agreement Notices indicated on the Signature Page.

 

9.7                                Licensee shall be solely responsible for determining if any tax on royalty income is owed outside the United States and shall pay the tax and be responsible for all filings with appropriate agencies of foreign governments.

 

9.8                                Additional royalties may be assessed by PHS on any payment that is more than ninety (90) days overdue at the rate of one percent (1%) per month.  This one percent (1%) per month rate may be applied retroactively from the original due date until the date of receipt by PHS of the overdue payment and additional royalties.  The payment of any additional royalties shall not prevent PHS from exercising any other rights it may have as a consequence of the lateness of any payment.

 

9.9                                All plans and reports required by this Article 9 and marked “confidential” by Licensee shall, to the extent permitted by law, be treated by PHS as commercial and financial information obtained from a person and as privileged and confidential, and any proposed disclosure of these records by the PHS under the Freedom of Information Act (FOIA), 5 U.S.C. §552 shall be subject to the predisclosure notification requirements of 45 C.F.R. §5.65(d).

 



 

9.10                         In the event PHS receives a license application from a Third Party Applicant for commercial development of one or more Licensed Products or Licensed Processes in the exclusive Licensed Fields of Use , as they pertain to Licensed Patent Rights for which the proposed commercial development is not specifically addressed in Licensee ’s then-current Commercial Development Plan (“ Third Party Applications ”), PHS shall notify Licensee , in writing, of the existence of the Third Party Applicant ’s license application.  Upon receipt of the written notice, Licensee shall respond in writing by either: (a) amending its Commercial Development Plan within one hundred and twenty (120) days in a manner acceptable to PHS to include a clinical research and development program for the proposed commercial development of the Third Party Applications including revised Benchmarks to be incorporated into Appendix E, and acceptance of the amendment to the Commercial Development Plan by PHS shall take into account if Licensee has already carried out work in respect of such Third Party Applications prior to notification by PHS ; or (b) amending its Commercial Development Plan within one-hundred eighty (180) days (or such longer period agreed by Licensee and such Third Party Applicant ) in a manner acceptable to PHS to include a joint pre-clinical research and development program with the Third Party Applicant for the proposed commercial development of the Third Party Applications ; or (c) granting an exclusive or non-exclusive sublicense under commercially reasonable terms to the Third Party Applicant under Licensed Patent Rights in respect of the Third Party Applications within one-hundred eighty (180) days (or such longer period agreed by Licensee and such Third Party Applicant ); or both (b) and (c).  If Licensee does not respond to the written notice as described in this Paragraph 9.10, and after thirty (30) days of final notice being sent to Licensee , PHS may remove the Licensed Products or Licensed Processes in respect of the Third Party Applications from the exclusive Licensed Field of Use in this Agreement , and PHS shall be free to grant a license to the Third Party Applicant under the Licensed Patent Rights in respect of the Third Party Applications .

 

10.                                PERFORMANCE

 

10.1                         Licensee shall use its reasonable commercial efforts to bring the Licensed Products and Licensed Processes to Practical Application .  “Reasonable commercial efforts” for the purposes of this provision shall include adherence to the Commercial Development Plan in Appendix E and performance of the Benchmarks in Appendix D.  The efforts of a sublicensee shall be considered the efforts of Licensee .

 

10.2                         Upon the First Commercial Sale , until the expiration or termination of this Agreement , Licensee shall use its reasonable commercial efforts to make Licensed Products and Licensed Processes reasonably accessible to the United States public.

 

10.3                         Licensee agrees, after its First Commercial Sale , to make reasonable quantities of Licensed Products or materials produced through the use of Licensed Processes available to patient assistance programs at cost.  Patient assistance programs are programs run by pharmaceutical companies to provide free medications to people who cannot afford to buy their medicine.  For each indication in each calendar year, the quantity of Licensed Products to be made available under this provision available to patient assistance programs at cost shall be defined as the higher of: (i) the maximum quantity of Licensed Products for such indication that was available in the previous calendar year (whether or not such Licensed Products were actually supplied); and (ii) five (5) percent of the total number of Licensed Products for such indication prescribed within the United States and its dependant territories in the previous calendar year.

 

10.4                         Licensee agrees, after its First Commercial Sale in a country in the Licensed Territory and as part of its marketing and product promotion in such country, to develop educational materials (e.g., brochures, website, etc.) directed to patients and physicians in that country detailing the Licensed

 



 

Products or medical aspects of the prophylactic and therapeutic uses of the Licensed Products to the extent permitted by law in such country.

 

10.5                         Licensee agrees to supply, upon request, to the Mailing Address for Agreement Notices indicated on the Signature Page, the Office of Technology Transfer, NIH with inert samples of the Licensed Products or Licensed Processes or their packaging for educational and display purposes only

 

11.                                INFRINGEMENT AND PATENT ENFORCEMENT

 

11.1                         PHS and Licensee agree to notify each other promptly of each infringement or possible infringement of the Licensed Patent Rights , as well as, any facts which may materially affect the validity, scope, or enforceability of the Licensed Patent Rights of which either party becomes aware.

 

11.2                         Pursuant to this Agreement and the provisions of 35 U.S.C. Part 29.  Licensee may in accordance with the provisions of Paragraph 11.3:

 

(a)                                  bring suit in its own name, at its own expense, and on its own behalf for infringement of presumably valid claims in the Licensed Patent Rights ;

 

(b)                                  in any suit, enjoin infringement and collect for its use, damages, profits, and awards of whatever nature recoverable for the infringement; or

 

(c)                                   settle any claim or suit for infringement of the Licensed Patent Rights .provided, however, that PHS and appropriate Government authorities shall have the first right to take such actions.

 

11.3                         If Licensee desires to initiate a suit for patent infringement, Licensee shall notify PHS in writing.  If PHS does not notify Licensee of its intent to pursue legal action within ninety (90) days, Licensee shall be free to initiate suit.  PHS shall have a continuing right to intervene in the suit.  Licensee shall take no action to compel the Government either to initiate or to join in any suit for patent infringement.  Licensee may request the Government to initiate or join in any suit if necessary to avoid dismissal of the suit Should the Government be made a party to any suit by motion or any other action of Licensee , Licensee shall reimburse the Government for any costs, expenses, or fees which the Government incurs as a result of the motion or other action.  In all cases, Licensee agrees to keep PHS reasonably apprised of the status and progress of any litigation.  Before Licensee commences an infringement action, Licensee shall notify PHS and give careful consideration to the views of PHS and to any potential effects of the litigation on the public health in deciding whether to bring suit.

 

11.4                         In the event that a declaratory judgment action alleging invalidity or non-infringement of any of the Licensed Patent Rights shall be brought against Licensee or raised by way of counterclaim or affirmative defense in an infringement suit brought by Licensee under Paragraph 11.3, pursuant to this Agreement and the provisions of 35 U.S.C. Part 29 or other statutes, Licensee may:

 

(a)                                  defend the suit in its own name, at its own expense, and on its own behalf for presumably valid claims in the Licensed Patent Rights ;

 

(b)                                  in any suit, ultimately to enjoin infringement and to collect for its use, damages, profits, and awards of whatever nature recoverable for the infringement; and

 



 

(c)                                   settle any claim or suit for declaratory judgment involving the Licensed Patent Rights -provided, however, that PHS and appropriate Government authorities shall have the first right to take these actions and shall have a continuing right to intervene in the suit; and

 

(d)                                  If PHS does not notify Licensee of its intent to respond to the legal action within a reasonable time, Licensee shall be free to do so.  Licensee shall take no action to compel the Government either to initiate or to join in any declaratory judgment action.  Licensee may request the Government to initiate or to join any suit if necessary to avoid dismissal of the suit.  Should the Government be made a party to any suit by motion or any other action of Licensee , Licensee shall reimburse the Government for any costs, expenses, or fees, which the Government incurs as a result of the motion or other action.  If Licensee elects not to defend against the declaratory judgment action, PHS , at its option, may do so at its own expense.  In all cases, Licensee agrees to keep PHS reasonably apprised of the status and progress of any litigation.  Before Licensee commences an infringement action, Licensee shall notify PHS and give careful consideration to the views of PHS and to any potential effects of the litigation on the public health in deciding whether to bring suit.

 

11.5                         In any action under Paragraphs 11.2, 11.3 or 11.4 the expenses including costs, fees, attorney fees, and disbursements, shall be paid by Licensee .  The value of any recovery made by Licensee through court judgment or settlement shall be treated as Net Sales and subject to earned royalties.

 

11.6                         PHS shall cooperate fully with Licensee in connection with any action under Paragraphs 11.2, 11.3 or 11.4.  PHS agrees promptly to provide access to all necessary documents and to render reasonable assistance in response to a request by Licensee .

 

12.                                NEGATION OF WARRANTIES AND INDEMNIFICATION

 

12.1                         PHS offers no warranties other than those specified in Article 1.

 

12.2                         PHS does not warrant the validity of the Licensed Patent Rights and makes no representations whatsoever with regard to the scope of the Licensed Patent Rights , or that the Licensed Patent Rights may be exploited without infringing other patents or other intellectual property rights of third parties.

 

12.3                         PHS MAKES NO WARRANTIES, EXPRESS OR IMPLIED, OF MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE OF ANY SUBJECT MATTER DEFINED BY THE CLAIMS OF THE LICENSED PATENT RIGHTS OR TANGIBLE MATERIALS RELATED THERETO.

 

12.4                         PHS does not represent that it shall commence legal actions against third parties infringing the Licensed Patent Rights .

 

12.5                         Licensee shall indemnify and hold PHS , its employees, students, fellows, agents, and consultants harmless from and against all liability, demands, damages, expenses, and losses, including but not limited to death, personal injury, illness, or property damage in connection with or arising out of:

 

(a)                                  the use by or on behalf of Licensee , its sublicensees, directors, employees, or third parties of any Licensed Patent Rights ; or

 

(b)                                  the design, manufacture, distribution, or use of any Licensed Products , Licensed Processes or materials by Licensee , or other products or processes developed in connection with or arising out of the Licensed Patent Rights .

 



 

12.6                         Licensee agrees to maintain a liability insurance program consistent with sound business practice.

 

13.                                TERM, TERMINATION, AND MODIFICATION OF RIGHTS

 

13.1                         This Agreement is effective when signed by all parties, unless the provisions of Paragraph 14.16 are not fulfilled, and shall extend to the expiration of the last to expire of the Licensed Patent Rights unless sooner terminated as provided in this Article 13.

 

13.2                         In the event that Licensee is in default in the performance of any material obligations under this Agreement , including but not limited to the obligations listed in Paragraph 13.5, and if the default has not been remedied within ninety (90) days after the date of notice in writing of the default, PHS may terminate this Agreement by written notice and pursue outstanding royalties owed through procedures provided by the Federal Debt Collection Act.

 

13.3                         In the event that Licensee becomes insolvent, files a petition in bankruptcy, has such a petition filed against it that is not discharged within ninety (90) days, determines to file a petition in bankruptcy, Licensee shall immediately notify PHS in writing.  Furthermore, PHS shall have the right to terminate this Agreement immediately upon Licensee ’s receipt of written notice.

 

13.4                         Licensee shall have a unilateral right to terminate this Agreement or any licenses in any country or territory by giving PHS sixty (60) days written notice to that effect.

 

13.5                         PHS shall specifically have the right to terminate or modify, at its option, this Agreement , if PHS determines that the Licensee :

 

(a)                                  is not executing the Commercial Development Plan submitted with its request for a license and the Licensee cannot otherwise demonstrate to PHS ’ satisfaction that the Licensee has taken, or can be expected to take within a reasonable time, effective steps to achieve Practical Application of the Licensed Products or Licensed Processes ;

 

(b)                                  has not achieved the Benchmarks as may be modified under Paragraph 9.2;

 

(c)                                   has willfully made a false statement of, or willfully omitted a material fact in the license application or in any report required by this Agreement ;

 

(d)                                  has committed a material breach of a covenant or agreement contained in this Agreement ;

 

(e)                                   is not keeping Licensed Products in a commercially reasonable manner available to the public after commercial use commences;

 

(f)                                    cannot reasonably satisfy unmet health and safety needs; or

 

(g)                                   cannot reasonably justify a failure to comply with the domestic production requirement of Paragraph 5.2 unless waived.

 

13.6                         In making the determination referenced in Paragraph 13.5, PHS shall take into account (a) the normal course of such commercial development programs relating to gene therapy conducted with sound and reasonable business practices and judgment, (b) regulatory considerations, and the annual reports submitted by Licensee under Paragraph 9.2.  Prior to invoking termination or modification of this Agreement under Paragraph 13.5, PHS shall give written notice to Licensee providing Licensee specific notice of, and a ninety (90) day opportunity to respond to, PHS ’ concerns as to the items referenced in 13.5(a)-13.5(g).  If Licensee fails to alleviate PHS ’ concerns as to the items

 



 

referenced in 13.5(a)-13.5(g) or fails to initiate corrective action to PHS ’ reasonable satisfaction, PHS may terminate this Agreement .

 

13.7                         When the public health and safety so require, and after written notice to Licensee providing Licensee a sixty (60) day opportunity to respond, PHS shall have the right to require Licensee to grant sublicenses to responsible applicants, on reasonable terms, in any Licensed Fields of Use under the Licensed Patent Rights , unless Licensee can reasonably demonstrate that the granting of the sublicense would not materially increase the availability to the public of the subject matter of the Licensed Patent Rights PHS shall not require the granting of a sublicense unless the responsible applicant has first negotiated in good faith with Licensee .

 

13.8                         PHS reserves the right according to 35 U.S.C. 5209(d)(3) to terminate or modify this Agreement if it is determined that this action is necessary to meet the requirements for public use specified by federal regulations issued after the date of the license and these requirements are not reasonably satisfied by Licensee .

 

13.9                         Within thirty (30) days of receipt of written notice of PHS ’ unilateral decision to modify or terminate this Agreement , Licensee may, consistent with the provisions of 37 C.F.R. §404.11, appeal the decision by written submission to the designated PHS official.  The decision of the designated PHS official shall be the final agency decision.  Licensee may thereafter exercise any and all administrative or judicial remedies that may be available.

 

13.10                  Within ninety (90) days of expiration or termination of this Agreement under this Article 13, a final report shall be submitted by Licensee .  Any royalty payments, including those incurred but not yet paid (such as the full minimum annual royalty), and those related to patent expense, due to PHS shall become immediately due and payable upon termination or expiration.  If terminated under this Article 13, sublicensees may elect to convert their sublicenses to direct licenses with PHS pursuant to Paragraph 4.3.  Unless otherwise specifically provided for under this Agreement , upon termination or expiration of this Agreement , Licensee shall return all Licensed Products or other materials included within the Licensed Patent Rights to PHS or provide PHS with certification of the destruction thereof.  Licensee may not be granted additional PHS licenses if the final reporting requirement is not fulfilled.

 

14.                                GENERAL PROVISIONS

 

14.1                         Neither party may waive or release any of its rights or interests in this Agreement except in writing.  The failure of a party to assert a right hereunder or to insist upon compliance with any term or condition of this Agreement shall not constitute a waiver of that right by that party or excuse a similar subsequent failure to perform any of these terms or conditions by the other party.

 

14.2                         This Agreement constitutes the entire agreement between the parties relating to the subject matter of the Licensed Patent Rights , Licensed Products and Licensed Processes , and all prior negotiations, representations, agreements, and understandings are merged into, extinguished by, and completely expressed by this Agreement .

 

14.3                         The provisions of this Agreement are severable, and in the event that any provision of this Agreement shall be determined to be invalid or unenforceable under any controlling body of law, this determination shall not in any way affect the validity or enforceability of the remaining provisions of this Agreement .

 

14.4                         If either party desires a modification to this Agreement , the parties shall, upon reasonable notice of the proposed modification by the party desiring the change, confer in good faith to determine the

 



 

desirability of the modification.  No modification shall be effective until a written amendment is signed by the signatories to this Agreement or their designees.

 

14.5                         The construction, validity, performance, and effect of this Agreement shall be governed by Federal law as applied by the Federal courts in the District of Columbia.

 

14.6                         All Agreement notices required or permitted by this Agreement shall be given by prepaid, first class, registered or certified mail or by an express/overnight delivery service provided by a commercial carrier, properly addressed to the other party at the address designated on the following Signature Page, or to another address as may be designated in writing by the other party.  Agreement notices shall be considered timely if the notices are received on or before the established deadline date or sent on or before the deadline date as verifiable by Postal Service postmark or dated receipt from a commercial carrier.  Parties should request a legibly dated Postal Service postmark or obtain a dated receipt from a commercial carrier or the Postal Service.  Private metered postmarks shall not be acceptable as proof of timely mailing.  Notices can also be sent by an email, or a fax.

 

14.7                         This Agreement shall not be assigned or otherwise transferred (including any transfer by legal process or by operation of law, and any transfer in bankruptcy or insolvency, or in any other compulsory procedure or order of court) except to Licensee’s Affiliate(s)  without the prior written consent of PHS .  The parties agree that the identity of the parties is material to the formation of this Agreement and that the obligations under this Agreement are nondelegable.  In the event that PHS approves a proposed assignment, Licensee shall pay PHS , as an additional royalty, one percent (1%) of the fair market value of any consideration received for any assignment of this Agreement within sixty (60) days of the assignment.

 

14.8                         Licensee agrees in its use of any PHS -supplied materials to comply with all applicable statutes, regulations, and guidelines, including PHS and HHS regulations and guidelines.  Licensee agrees not to use the materials for research involving human subjects or clinical trials in the United States without complying with 21 C.F.R. Part 50 and 45 C.F.R. Part 46.  Licensee agrees not to use the materials for research involving human subjects or clinical trials outside of the United States without notifying PHS , in writing, of the research or trials and complying with the applicable regulations of the appropriate national control authorities.  Written notification to PHS of research involving human subjects or clinical trials outside of the United States shall be given no later than sixty (60) days prior to commencement of the research or trials.

 

14.9                         Licensee acknowledges that it is subject to and agrees to abide by the United States laws and regulations (including the Export Administration Act of 1979 and Arms Export Control Act) controlling the export of technical data, computer software, laboratory prototypes, biological material, and other commodities.  The transfer of these items may require a license from the appropriate agency of the U.S. Government or written assurances by Licensee that it shall not export these items to certain foreign countries without prior approval of this agency.  PHS neither represents that a license is or is not required or that, if required, it shall be issued.

 

14.10                  To the extent practicable and allowed by law and regulation, Licensee agrees to mark the Licensed Products or their packaging sold in the United States with all applicable U.S. patent numbers and similarly to indicate “Patent Pending” status.  All Licensed Products manufactured in, shipped to, or sold in other countries shall be, to the extent practicable and allowed by law and regulation in such countries, marked in a manner to preserve PHS patent rights in those countries.

 

14.11                  By entering into this Agreement , PHS does not directly or indirectly endorse any product or service provided, or to be provided, by Licensee whether directly or indirectly related to this Agreement Licensee shall not state or imply that this Agreement is an endorsement by the Government, PHS ,

 



 

any other Government organizational unit, or any Government employee.  Additionally, Licensee shall not use the names of NIH , FDA , PHS , or HHS or the Government or their employees in any advertising, promotional, or sales literature without the prior written approval of PHS .

 

14.12                  The parties agree to attempt to settle amicably any controversy or claim arising under this Agreement or a breach of this Agreement , except for appeals of modifications or termination decisions provided for in Article 13.  Licensee agrees first to appeal any unsettled claims or controversies to the designated PHS official, or designee, whose decision shall be considered the final agency decision.  Thereafter, Licensee may exercise any administrative or judicial remedies that may be available.

 

14.13                  Nothing relating to the grant of a license, nor the grant itself, shall be construed to confer upon any person any immunity from or defenses under the antitrust laws or from a charge of patent misuse, and the acquisition and use of rights pursuant to 37 C.F.R. Part 404 shall not be immunized from the operation of state or Federal law by reason of the source of the grant.

 

14.14                  Any formal recordation of this Agreement required by the laws of any Licensed Territory as a prerequisite to enforceability of the Agreement in the courts of any foreign jurisdiction or for other reasons shall be carried out by Licensee at its expense, and appropriately verified proof of recordation shall be promptly furnished to PHS .

 

14.15                  Paragraphs 4.3, 8.1, 9.5-9.7 (in respects of sales carried out prior to termination), 12.1-12.4, 12.5 (in respects of acts carried out prior to termination), 13.9, 13.10, 14.12 and 14.15 of this Agreement shall survive termination of this Agreement .

 

14.16                  The terms and conditions of this Agreement shall, at PHS ’ sole option, be considered by PHS to be withdrawn from Licensee ’s consideration and the terms and conditions of this Agreement , and the Agreement itself to be null and void, unless this Agreement is executed by the Licensee and a fully executed original is received by PHS within sixty (60) days from the date of PHS signature found at the Signature Page.

 

SIGNATURES BEGIN ON NEXT PAGE

 



 

PHS PATENT LICENSE AGREEMENT - EXCLUS1VE

 

SIGNATURE PAGE

 

For PHS :

 

/s/ Richard U. Rodriquez

 

8-5-11

Richard U. Rodriguez

 

Date

Director, Division of Technology Development and Transfer

 

 

Office of Technology Transfer

 

 

National Institutes of Health

 

 

 

Mailing Address or E-mail Address for Agreement notices and reports:

 

Chief, Monitoring & Enforcement Branch
Office of Technology Transfer
National Institutes of Health
6011 Executive Boulevard, Suite 325
Rockville, Maryland 20852-3804 U.S.A.

 

E-mail:  LicenseNotices_ Reports@mail.nih.gov

 

For Licensee (Upon, information and belief, the undersigned expressly certifies or affirms that the contents of any statements of Licensee made or referred to in this document are truthful and accurate.)

 

By:

 

 

 

 

 

 

 

 

/s/ Piers Morgan

 

10 August 2011

 

 

 

Signature of Authorized Official

 

Date

 

Piers Morgan

 

Chief Financial Officer

 

Amsterdam Molecular Therapeutics

 



 

Mailing Address for Agreement notices:

 

Chief Executive Officer

 

Amsterdam Molecular Therapeutics

 

P.O. Box 22506

 

1100 DA Amsterdam

 

The Netherlands

 

Tel. +31(0)20 566 7394

 

I.  Official and Mailing Address for Financial notices ( Licensee’s contact person for royalty payments)

 

Piers Morgan

 

Chief Financial Officer

 

Amsterdam Molecular Therapeutics

 

P.O. Box 22506

 

1100 DA Amsterdam

 

The Netherlands

 

Tel.+31(0)20 566 7394

 

E-mail: p.morgan@amtbiopharma.com

 

Any false or misleading statements made, presented, or submitted to the Government , including any relevant omissions, under this Agreement and during the course of negotiation of this Agreement are subject to all applicable civil and criminal statutes including Federal statutes 31 U.S.C. §§3801-3812 (civil liability) and 18 U.S.C. §1001 (criminal liability including fine(s) or imprisonment).

 



 

APPENDIX A - PATENT(S) OR PATENT APPLICATION(S)

 

Patent(s) or Patent Application(s):

 

1. U. S. Patent 6,984,517, issued January 10, 2006, entitled “AAV5 Vector and Uses Thereof; U.S. Patent 7,479,554, issued January 20, 2009, entitled “AA V5 Nucleic Acids”as well as all issued and pending foreign counterparts [ HHS Ref. No. E-127-1998/0];

 

2. U. S. Patent 6,855,314, issued February 15, 2005, entitled “AAV5 Vector for Transducing Brain Cells and Lung Cells” [HHS Ref. No. E-072-2000/0].

 



 

APPENDIX B - LICENSED FIELDS OF USE AND TERRITORY

 

I.                                         Licensed Fields of Use :

 

(a)                                  Exclusive Licensed Field of Use : (i) Use of the Licensed Patent Rights for the development and sale of AAV5 based therapeutic products to be delivered to the brain or liver for treatment of human diseases originating in the brain or liver; (ii) Note that arthritis related diseases are expressly excluded.

 

(b)                                  Non-Exclusive Licensed Field of Use : Use of the Licensed Patent Rights for the development and sale of AAV5 based therapeutic products to treat human diseases other than the ones covered under (a)(i).

 

II.                                    Licensed Territory :

 

(a)                                  Worldwide.

 



 

APPENDIX C - ROYALTIES

 

Royalties:

 

I.                                         Licensee agrees to pay to PHS a noncreditable, nonrefundable license issue royalty in the amount of one hundred forty thousand dollars ($140,000).  Payment will be made in two tranches, the first payment of one hundred thousand dollars ($100,000) being payable within sixty (60) days from the effective date of this Agreement ; the second payment of forty thousand dollars ($40,000) being payable on March 1, 2012.  If this Agreement is terminated by Licensee on or before March 1, 2012, Licensee agrees to pay the remaining tranch of license issue royalty in full within sixty (60) days before termination

 

II.                                    Licensee agrees to pay to PHS a nonrefundable minimum annual royalty in the amount of fifteen thousand dollars ($15,000) as follows:

 

(a)                                  The first minimum annual royalty is due within sixty (60) days of the effective date of this Agreement and may be prorated according to the fraction of the calendar year remaining between the effective date of this Agreement and the next subsequent January 1; and

 

(b)                                  Subsequent minimum annual royalty payments are due and payable on January 1 of each calendar year and may be credited against any earned royalties due for sales made in that year.

 

III.                               Licensee agrees to pay PHS earned royalties of one and two-tenth percent (1.2%) on Net Sales by or on behalf of Licensee and its sublicensees.

 

IV.                                Licensee agrees to pay PHS Benchmark royalties within sixty (60) days of achieving each Benchmark :

 

(a)                                  Thirty-one thousand and five hundred U.S. dollars ($31,500) - Initiation of each Phase 1 clinical trial or foreign equivalent.

 

(b)                                  Seventy-eight thousand five hundred U.S. dollars ($78,500) - Initiation of each Phase II clinical trial or foreign equivalent.

 

(c)                                   One hundred and fifty-seven thousand and five hundred U.S. dollars ($157,500) - Initiation of each Phase III clinical trial or foreign equivalent.

 

(d)                                  Initiation of first Marketing Approval or foreign equivalent for any indications in the liver in the following jurisdictions/countries:

 

(i)                                      Three hundred fifteen thousand U.S. dollars ($315,000) in Europe.

 

(ii)                                   Three hundred fifteen thousand U.S. dollars ($315,000) in the United States.

 

(iii)                                Seventy-eight thousand five hundred U.S. dollars ($78,500) in Australia.

 

(iv)                               Seventy-eight thousand five hundred U.S. dollars ($78,500) in Canada.

 

(v)                                  Seventy-eight thousand five hundred U.S. dollars ($78,500) in Japan.

 

(e)                                   Initiation of first Marketing Approval or foreign equivalent for any indications in the brain in the following jurisdictions/countries:

 

(i)                                      Three hundred fifteen thousand U.S. dollars (S315,000) in Europe.

 



 

(ii)                                   Three hundred fifteen thousand U.S. dollars ($315,000) in the United States.

 

(iii)                                Seventy-eight thousand five hundred U.S. dollars ($78,500) in Australia.

 

(iv)                               Seventy-eight thousand five hundred U.S. dollars ($78,500) in Canada.

 

(v)                                  Seventy-eight thousand five hundred U.S. dollars ($78,500) in Japan.

 

V.                                     Licensee agrees to pay PHS additional sublicensing royalties, as following, on the fair market value of any consideration received for granting each sublicense within sixty (60) days of the execution of each sublicense:

 

(a)                                  For any sublicense executed by the Licensee before the completion of any phase I clinical trial or foreign equivalent for any disease indications, Licensee agrees to pay a sublicensing royalty of ten percent (10%); and

 

(b)                                  For any sublicense executed by the Licensee after the completion of any phase I clinical trial or foreign equivalent but before the completion of any phase II clinical trial or foreign equivalent for any disease indications, Licensee agrees to pay a sublicensing royalty of six percent (6%); and

 

(c)                                   For any sublicense executed by the Licensee either after the completion of any phase II clinical trial or foreign equivalent for any disease of Orphan Indication , or after the completion of any phase III clinical trial or foreign equivalent for any other disease indications, whichever comes first.  Licensee agrees to pay a sublicensing royalty of three percent (3%).

 

Contractual payments made by a sublicensee to the Licensee or an Affiliate received after the effective date of this Agreement for costs, services and expenses for the Licensee or Affiliate to conduct, supervise or participate in one or more clinical trial(s) for the development of the Licensed Products shall not be accounted for as sublicensing royalties.

 



 

APPENDIX D - BENCHMARKS AND PERFORMANCE

 

Licensee agrees to the following Benchmarks for its performance under this Agreement and, within thirty (30) days of achieving a Benchmark , shall notify PHS that the Benchmark has been achieved.

 

Benchmarks for Licensed Products of Orphan Indication (there is no formal Phase III clinical trial required for Marketing Approval) - liver

 

I.                                         Initiation of first Phase I clinical trial or foreign equivalent - 2012

 

II.                                    Initiation of first Phase II clinical trial or foreign equivalent - 2014

 

III.                               Submission to Regulatory Authority of first Marketing Approval or foreign equivalent - 2015

 

Benchmarks for Licensed Products - brain

 

I.                                         Initiation of Preclinical Development phase or foreign equivalent - 2011

 

II.                                    Initiation of first Phase I clinical trial or foreign equivalent - 2012

 

III.                               Initiation of first Phase II clinical trial or foreign equivalent - 2014

 

IV.                                Initiation of first Phase III clinical trial or foreign equivalent - 2017

 

V.                                     Submission to Regulatory Authority of first Marketing Approval or foreign equivalent - 2020

 



 

APPENDIX E - COMMERCIAL DEVELOPMENT PLAN

 

Project Plan Details - Liver:

 

Acute intermittent porphyria (AIP) is an autosomal dominant inherited condition caused by mutations in the porphobilinogen deaminase (PBGD) gene.  The PBGD gene is located on chromosome 11q24.1-24.2 and spread over fifteen exons.  The protein encoded by this gene is a rate-limiting enzyme, the PBGD enzyme, in the haem synthetic pathway.

 

More than 225 mutations of the PBGD gene have been described, all of them associated with loss of catalytic function.  The disease shows incomplete penetrance and only 20-50% of persons with one or more of the described mutations exhibit clinical symptoms of the disease.  The genetic disorder results in a 50% reduction of PBGD enzymatic activity.  This reduction of hepatic PBGD activity leads to an accumulation of toxic metabolites resulting from the blockade within the haem synthesis pathway.  Concentrations of haem precursors porphobilinogen (PGB) and delta-aminolevulinic acid (ALA) increase in blood and urine.  Lack of haem and/or accumulation of these metabolites are responsible for the acute attacks characteristic of this disease (Kauppinen et al 2005; Herrick and McColl 2005).  Currently, there is no treatment available for the disease.

 

Over the last couple of years Licensee has explored AMT-021 (replication defective recombinant adeno-associated viral vector, AAV, containing the porphobilinogen deaminase gene) for therapeutic intervention in AIP.  AMT-021 is an AAV with pseudotype 5 capsid, which expresses the human PBGD gene under the transcriptional control of a liver specific promoter.  The therapeutic expression cassette consists of the human PBGD cDNA (codon optimised for human expression) inserted downstream of the liver specific promoter EalbAAT and upstream of a human PBGD polyadenylation sequence.

 

AMT-021 acts by delivering the PBGD expression cassette directly into hepatocytes.  The increase of PBGD enzymatic activity in the liver of AIP patients will provide sufficient enzyme to prevent the accumulation of toxic metabolites and thus, prevent porphyric attacks.

 

The aim of the project is to bring AAV5-PBDG therapy to patients.  Licensee has already secured orphan designation for AAV5-PBDG treatment for AIP in Europe.  The table below describes the outline development plans, starting from a research batch production, and moving through to primate proof-of-concept, tox batch, pre-observational study, product development, GMP production, Phase I/II clinical trial, Phase Il/III clinical trial, all the way to regulatory filing.  Please note that the timelines are preliminary only, and that it is the nature of scientific and clinical development that planned timelines may change.

 

The aim of this project is to develop a gene therapy product for the treatment of AIP, and to deliver a data package that is suitable for the submission and approval by the European and North American regulatory authorities.

 

Vector development and manufacturing


To develop a gene therapy for PBGD deficient patients, AAV5-PBDG product was designed to expresses the human PBGD gene under the control of a liver specific promoter.  AAV5-PBDG was produced in insect cells using the recombinant baculovirus method; sufficient amount of material was produced for efficacy studies in mice.  Methods to determine the quantity and purity of the rAAV batches were developed.  A purification process including chromatography and filtration steps was developed, further optimization and characterization of the scale-up procedure will be performed before a final batch for toxicology, for proof of principle and for clinical trials can be produced.

 

PoC in pre-clinical models


Because total deficiency of PBGD is lethal in mice, a compound heterozygous mouse (PBGD+/- referred to as AIP mice) with ~35% of normal hepatic PBGD activity, has been developed as an established model to study AIP.  This murine model of AIP exhibits, after disease induction with phenobarbital (Pb), the typical biochemical characteristics

 



 

of human AIP, notably, decreased hepatic PBGD activity, massively increased urinary excretion of haem precursors (ALA and PBG) and decreased motor function.

 

AIP mice were used to test the AAV5-PBDG product.  The therapeutic effect was evaluated three month after a single intravenous administration of AAV5-PBDG.  Efficacy of the therapy was demonstrated as the treatment was able to prevent disease induction with Pb.  ALA and PBG levels in treated animals was reduced, and motor disturbance induced by Pb treatment, as measured in the Rotarod test, was almost completely abolished.  In addition, PBGD enzymatic activity increased in the AAV5-PBDG treated group 10 times over that of the control group.

 

This initial PoC will be repeated with the final version of the therapeutic vector following the completion of the vector development and manufacturing optimization.  The final PoC will include the following:

 

PoC in rodent disease model

 

·                   PoC in non-human primates, based on agreed protocol

 

GLP Toxicology

 

The aim of this section is to deliver toxicology study report suitable for the submission the regulatory authority.  The work will entail the following:

 

·                   Scientific advice from a regulatory body (AEMPS and/or EMA) for safety and toxicology package

·                   GLP toxicology study in rodents rats or mice, including any required biodistribution studies

·                   Supportive data for toxicology study in non-human primates

·                   GLP germline transmission study

 

Toxicology study design will take into account:

 

·                   Identification of potential target organs of biological activity and of potential target organs of toxicity

·                   Eventual concomitant medication (e.g. immunosuppressants, standard co-medication)

·                   Environmental risk/shedding

·                   Analysis of appropriateness of surrogate markers of efficacy/safety

·                   Any other relevant issues as may be identified

 

Clinical observational, pre-intervention study/studies

 

Before entering the interventional clinical study, an observation clinical study will be conducted to provide baseline information on the course of the disease by recording episodes AIP, abdominal pain, hospitalizations, extent of any possible known or unknown to be related to AIP symptomatology, incidence of (adverse) clinical events per year, etc.  Sufficient data will be collected to provide a clinical picture to obtain a baseline data and to determine how efficacy will be shown during the interventional clinical trial.

 

Phase I/II

 

The clinical phase I/II should include an estimated minimum 6 patients that are administered the gene therapy drug, and are followed up and clinically assessed for at least 6 months following drug administration.  The primary aim of the clinical study will be safety and efficacy of the AAV5-PBDG product.  The clinical trial will include all biochemical, imaging, clinical and functional assays to assess the disease state and change therein over time, the phenotypic disease variation, as well as the overall clinical and psychosocial or other health status or change therein over time of the individual trial subjects, both before, during and following drug administration.

 

Phase II/III & Regulatory submission

 

After successful completion of Phase I/Il study a Phase II/III trial will be conducted with the aim of bringing the AIP therapy to market.  Licensee estimates that 20-30 patients in total would be sufficient for regulatory filing of this product, as AIP is an ultra-orphan disease with a very limited patient number world-wide.

 



 

Project Plan Details-Brain (Parkinson’s Disease)

 

Parkinson’s disease (PD) is a progressive neurodegenerative disease, resulting in tremors, stiffness, slowness of movement, and lack of coordination.  Patients are faced with a severely debilitating disease and a serious loss in quality of life.  PD is caused by degeneration and death of nerve cells in a specific part of the brain known as the substantia nigra.  These cells produce dopamine, a substance necessary for communication between nerve cells involved in the coordination of movement.

 

PD is the second most common neurodegenerative disease.  It usually affects people over 65, with an estimated total of 4.5 million patients worldwide.  Due to increasing life expectancy of the general population, the number of patients with PD is expected to double to around 9 million patients between now and 2030.

 

An ideal therapy for PD would decrease disability and slow down or halt disease progression.  Unfortunately, such treatments are not available yet and current therapies are limited to symptomatic treatment only.  These include levodopa, dopamine agonists, monoamine oxidase B (MAO-B) inhibitors and anticholinergic agents.

 

Glial cell line-derived neurotrophic factor (GDNF) was shown to promote the survival and differentiation of dopaminergic neurons.  The therapy aims to protect and enhance the function of the dopamine-producing nerve cells in the brain.  To date a number of clinical trials have been conducted in which recombinant GDNF protein has been directly delivered to the PD brain, using a delivery pump device implanted into patients’ abdomen.  Although the results were inconsistent, due to the difficulty of delivering protein continuously into the brain via an implanted pump, some patients have shown a significant clinical response to the treatment.  It is therefore not a question whether this approach works, because it definitely did in some patients, but rather how it can be done more consistently.  AAV-GDNF gene therapy treatment would result in continues delivery of GDNF protein into brain, and is therefore likely to result in significant clinical benefit for PD patients.

 

Licensee has recently started preclinical development of AAV-GDNF gene therapy that will introduce the gene coding for GDNF using recombinant adeno associated virus vector (AAV).  AAV serotype 5 has been shown to be the serotype of choice for gene delivery into the brain.  After successful proof of concept (POC) and toxicology studies in rodents and primates, AMT will start an extensive clinical development.

 

The aim of this project is to develop a gene therapy product for the treatment of Parkinson’s disease, and to deliver a data package that is suitable for the submission and approval by the European and North American regulatory authorities.

 

Vector development and manufacturing

 

To develop a gene therapy for Parkinson’s disease, AAV-GDNF product was designed to expresses the human GDNF and is produced in insect cells using the recombinant baculovirus method.  The AAV5-GDNF is based on Licensee ’s standard manufacturing process, but in addition incorporates recent new technology of the basic process and makes use of an optimized Rep baculovirus construct in the upstream process and an additional chromatography step in the downstream process.  This optimisation delivers enhanced quality and robustness of the AAV5-GDNF product.  This process is fully scalable and allows for manufacturing of sufficient GMP-compliant product for PD patients.

 

Characterization of AAV5-GDNF

 

The AAV5-GDNF was tested in a functional in vitro assay in cultured E13.5 rat DRG explants.  Vigorous neural outgrowth was observed, indicating that the produced AAV5-GDNF is capable of mediating secretion of biologically functional recombinant GDNF.

 

In vivo characterization

 

Subsequently, an in-vivo characterisation of the AAV5-GDNF has been conducted.  Three different concentrations of AAV5-GDNF were injected unilaterally into the rat striatum.  Brains were analyzed for GDNF expression 6 weeks post injection using immunohistochemistry.  Resulting data demonstrated that there is a strong, concentration dependent GDNF expression throughout the injected hemisphere.

 



 

PoC in pre-clinical models

 

The produced AAV5-GDNF will be used to show biological activity and efficacy in animal models of Parkinson’s disease.  These experiments will be conducted using rat models of Parkinson’s disease (in collaboration with University of Lund, Sweden) as well as non-human primates’ model of Parkinson’s disease (in collaboration with CEA, Paris, France).  In addition to distribution studies, onset and kinetics of GDNF expression, neurochemical measurements (dopamine and dopamine metabolites), immunohistochemistry and behavioral studies will be conducted to test for functional improvement.

 

GLP Toxicology

 

The definitive design of the actual studies will be finalized after discussions with relevant agencies.  Licensee proposes to conduct a six months study in mice and in parallel a 6-12 months study in non-human primates to account for the safety of the drug.  The studies will comprise four test groups: 1.  Control (vehicle), 2.  Low dose (No observed effect level (NOEL) in the proof-of concept studies), 3.  Mid-dose (highest dose considered for clinical studies), and 4.  High dose (10 times higher than the mid-dose).

 

The protocol will include the following evaluations:

 

·                   Clinical Signs: recorded daily, beginning 7 days prior to surgery

·                   Food Consumption: recorded daily, beginning 7 days prior to surgery

·                   Body Weight: Once pre-surgery, day of surgery, then bi-weekly

·                   Clinical Chemistry: Twice a month presurgery, one week post surgery, then monthly

·                   Hematology: Twice a month presurgery, one week post surgery, then monthly

·                   Coagulation: Twice a month presurgery, one week post surgery, then monthly

·                   Antibodies against GDNF or AAV5 in plasma, twice prior to surgery, monthly thereafter.

·                   PK - CSF: To determine if there is GDNF in the CSF, twice prior to surgery, monthly thereafter.

·                   Neurological Examination: Twice prior to surgery, Day 7 post surgery, monthly thereafter

·                   MRI (T1,T2): Once prior to surgery, within three hours post surgery, and within three days prior to necropsy.

·                   Pathology

1. Gross pathology at necropsy

2. Selected peripheral tissues collected for histopathological analysis by a Board Certified Pathologist

3. Complete CNS histopathological assessment by a Board Certified Neuropathologist, peer reviewed by another Board Certified Pathologist

·                   Q-PCR in selected organs in order to assess any biodistribution of the vector DNA to other organs.

 

Phase I/II

 

The primary objective of the clinical phase I/II will be to assess the safety and feasibility of intra-putaminal delivery of AAV5-GDNF to patients with PD.  Secondary objectives include measuring clinical efficacy and demonstrating improvement in a surrogate marker end point (18F-Dopa PET) as proof of concept.

 

Licensee is proposing a single centre open label trial of striatally delivered AAV5-GDNF in PD employing a dose escalation design to assess the mentioned primary and secondary outcome measures.  Licensee anticipates enrolling 12 patients in this study, with an escalating dose group design with three patients in each dose group.  Licensee will start with the lowest dose and progress in an incremental way to higher doses.

 

Primary outcome assessments will be performed at two weeks, one month, three months, six months, 12 months and 18 months post intra-putaminal infusion of AAV5-GDNF.  Clinical secondary outcome assessments will be performed at three months, six months, 12 months and 18 months post intra-putaminal infusion of AAV5-GDNF.  18F-dopa PET secondary outcome assessments will be performed at six months and 18 months post intra-putaminal infusion of AAV5-GDNF.

 

If feasibility and safety is confirmed and, serial PET imaging demonstrates increased 18F-dopa uptake with a trend towards clinical improvement, we will proceed to phase 2/3 clinical trials.

 



 

Phase II/III, Phase III & Regulatory submission

 

After successful completion of Phase I/II study, two additional clinical trials will be required.  The final plans for these trials will be optimized based on the outcome of the Phase I/II study.  Licensee estimates 50 patients to be enrolled in the Phase II/III clinical study, and 500 patients to be enrolled in the pivotal trial, the details however will be established, based on the outcome of the Phase I/II trial.

 

Additional indication;

 

In addition to the above, Licensee has an active programs in hemophilia B using AAV5-Factor IX, in hemophilia A using AAV5-Factor VIII, in Sanfilippo B - currently conducted by Institut Pasteur, using AAV5-NaGlu gene, and a program for the development of treatment for Usher syndrome type 1 (USH1) using AAV5-MY07A.  Additional early stage programs are under evaluation.

 



 

APPENDIX F - EXAMPLE ROYALTY REPORT

 

Required royalty report information includes:

 

·                   OTT license reference number (L-XXX-200X/0)

·                   Reporting period

·                   Catalog number and units sold of each Licensed Product (domestic and foreign)

·                   Gross Sales per catalog number per country

·                   Total Gross Sales

·                   Itemized deductions from Gross Sales

·                   Total Net Sales

·                   Earned Royalty Rate and associated calculations

·                   Gross Earned Royalty

·                   Adjustments for Minimum Annual Royalty (MAR) and other creditable payments made

·                   Net Earned Royalty due

 

Example

 

Catalog Number

 

Product Name

 

Country

 

Units Sold

 

Gross Sales
(US$)

 

1

 

A

 

US

 

250

 

62,500

 

1

 

A

 

UK

 

32

 

16,500

 

1

 

A

 

France

 

25

 

15,625

 

2

 

B

 

US

 

0

 

0

 

3

 

C

 

US

 

57

 

57,125

 

4

 

D

 

US

 

12

 

1,500

 

 

 

 

 

 

 

Total Gross Sales

 

153,250

 

 

 

 

 

 

 

Less Deductions:

 

 

 

 

 

 

 

 

 

Freight

 

3,000

 

 

 

 

 

 

 

Returns

 

7,000

 

 

 

 

 

 

 

Total Net Sales

 

143,250

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Royalty Rate

 

8

%

 

 

 

 

 

 

Royalty Due

 

11,460

 

 

 

 

 

 

 

Less Creditable Payments

 

10,000

 

 

 

 

 

 

 

Net Royalty Due

 

1,460

 

 



 

APPENDIX G - ROYALTY PAYMENT OPTIONS

 

The OTT License Number MUST appear on payments, reports and correspondence.

 

Automated Clearing House (ACH) for payments through U.S. banks only

 

The NIH encourages our licensees to submit electronic funds transfer payments through the Automated Clearing House (ACH).  Submit your ACH payment through the U.S. Treasury web site located at: https://www.pay.gov .  Locate the “NIH Agency Form” through the Pay.gov “Agency List”.

 

Electronic Funds Wire Transfers

 

The following account information is provided for wire payments.  In order to process payment via Electronic Funds Wire Transfer sender MUST supply the following information within the transmission:

 

Drawn on a U.S. bank account via FEDWIRE should be sent directly to the following account:

 

Beneficiary Account:

Federal Reserve Bank of New York or TREAS NYC

Bank:

Federal Reserve Bank of New York

ABA#

021030004

Account Number:

75080031

Bank Address:

33 Liberty Street, New York, NY 10045

Payment Details:

License Number (L-XXX-XXXX)

 

Name of Licensee

 

Drawn on a foreign bank account should be sent directly to the following account. Payment must be sent in U.S. Dollars (USD) using the following instructions:

 

Beneficiary Account:

Federal Reserve Bank of New York/ITS or FRBNY/ITS

Bank:

Citibank N.A. (New York)

SWIFT Code:

CITIUS33

Account Number:

36838868

Bank Address:

388 Greenwich Street, New York, NY 10013

Payment Details (Line 70):

NIH 75080031

 

License Number (L-XXX-XXXX)

 

Name of Licensee

Detail of Charges (line 71a):

Charge Our

 



 

Checks

 

All checks should be made payable to “NIH Patent Licensing”

 

Checks drawn on a U.S. bank account and sent by US Postal Service should be sent directly to the following address:

 

National Institutes of Health (NIH)

P.O. Box 979071

St. Louis, MO 63197-9000

 

Checks drawn on a U.S. bank account and sent by overnight or courier should be sent to the following address:

 

US Bank

Government Lockbox SL-MO-C2GL

1005 Convention Plaza

St. Louis, MO 63101

Phone: 314-418-4087

 

Checks drawn on a foreign bank account should be sent directly to the following address:

 

National Institutes of Health (NIH)

Office of Technology Transfer

Royalties Administration Unit

6011 Executive Boulevard

Suite 325, MSC 7660

Rockville, Maryland 20852

 



 

NATIONAL INSTITUTES OF HEALTH

 

FIRST AMENDMENT TO L-116-2011/0

 

This is the first amendment (“First Amendment”) of the agreement by and between the National Institutes of Health (“NIH”) within the Department of Health and Human Services (“HHS”), and UniQure biopharma B.V. (formerly Amsterdam Molecular Therapeutics N.V. (AMT)) having an effective date of August 10, 2011 and having NIH Reference Number L-l16-2011/0 (“Agreement”).  This First Amendment, having NIH Reference Number L-l16-2011/1, is made between the NIH through the Office of Technology Transfer, NIH, having an address at 6011 Executive Boulevard, Suite 325, Rockville, Maryland 20852-3804, U.S.A., and UniQure biopharma B.V. (formerly Amsterdam Molecular Therapeutics N.V. (AMT)), having an office at Meibergdreef 61, 1105 BA Amsterdam, The Netherlands (“ Licensee ”).  This First Amendment includes, in addition to the amendments made below, 1) a Signature Page, and 2) Attachment 1 (Royalty Payment Information).

 

WHEREAS, NIH and Licensee desire that the Agreement be amended a first lime as set forth below in order to

 

a)                                      Change the name of Licensee from Amsterdam Molecular Therapeutics N.V. (AMT) to UniQure biopharma B.V. (UniQure).  This name change is the result of a transaction that took place on 30 March 2012, whereby AMT, a public company, was liquidated and all its operations and stocks were transferred to UniQure, a privately held company.

 

b)                                      Modify language related to financial terms associated with sublicensing, so as to cause a reduction in financial obligations due to NIH from sublicensing of the Agreement by Licensee in order to expedite the development of therapeutics for rare diseases.

 

NOW, THEREFORE, in consideration of the mutual covenants and promises contained herein, NIH and Licensee, intending to be bound, hereby mutually agree to the following:

 

1)                                      a)                                      In Cover page following the list of “licensed patent and patent application”, the name of Licensee has been changed to UniQure biopharma B.V.

 

b)                                      In the signature page under “signature of authorized official”, under “mailing address for Agreement notices”, and under “official and mailing address for financial notices” “Amsterdam Molecular Therapeutics, N.V.” has been changed to UniQure biopharma B.V.

 

c)                                       In the caption of the Agreement AMT is changed to UniQure.

 

2)                                      Replace Paragraph 6.7 with the following:

 

6.7                                      No multiple royalties shall be payable if any Licensed Products or Licensed Processes are covered by more than one of the Licensed Patent Rights. In the event that this Agreement and NIH license L-107-2007/0 as amended from time to time apply to the same product sold by the Licensee or its sublicensees, then the Licensee shall only pay earned royalties, benchmark royalties, and sublicensing royalties under this Agreement.

 

3)                                      Replace Appendix C Section V with the following:

 

Licensee agrees to pay NIH additional sublicensing royalties, as follows, on the fair market value of any consideration received for granting each sublicense within sixty (60) days of the execution of each sublicense:

 



 

(i)             For any sublicense executed by the Licensee before the completion of any phase I clinical trial or foreign equivalent for any disease indication, Licensee agrees to pay a sublicensing royalty as in the following formula:

 

Sublicens ing Royalty = 10.0% x P/(P+T+L)

 

for the purposes of calculating sublicensing royalties in (i), where P/(P+T+L) is a fraction in which P represents the NIH ’s Licensed Patent Right , T represents the Intellectual Property (IP) licensed by Licensee from a third party, and where such an IP is related only to an active component of the Licensed Products (i.e. gene of interest incorporated into the AAV construct), and L represents Licensee’s own IP used to make the Licensed Product . Furthermore P, T and L, when present, each carries a value of 1.
The value of the fraction P/(P+T+L) as applied to (i) can never go below 1/3 (0.33), and therefore the Sublicensing Royalty as applied to (i) will never go below 3.3% (10.0% x 0.33).

 

(ii)            For any sublicense executed by the Licensee after the completion of any phase 1 clinical trial or foreign equivalent but before the completion of any phase II clinical trial or foreign equivalent for any disease indication, Licensee agrees to pay a sublicensing royalty as in the following formula:

 

Sublicensing Royalty =6.0% x P/(P+T+L)

 

The value of the fraction P/(P+T+L) as applied to (ii) can never go below 0.45, and therefore the Sublicensing Royalty as applied to (ii) will never go below 2.7% (6.0% x 0.45)

 

(iii)           For any sublicense executed by the Licensee either after the completion of any phase II clinical trial or foreign equivalent for any disease of Orphan Indication, or Ultra-Orphan Indication, or after the completion of any phase III clinical trial or foreign equivalent for any other disease indications, Licensee agrees to pay a sublicensing royalty as in the following formula:

 

Sublicensing Royalty = 3.0% x P/(P+T+L)

 

The value of the fraction P/(P+T+L) as applied to (iii) can never go below 0.65, and therefore the Sublicensing Royalty as applied to (iii) will never go below 1.95% (3.0% x 0.65)

 

Contractual payments made by a sublicensee to the Licensee or an Affiliate received after the effective date of this Agreement for costs, services and expenses for the Licensee or Affiliate to perform research and development activities, or to conduct, supervise or participate in one or more clinical trial(s) for the development of the Licensed Products, or to manufacture clinical and commercial batches of Licensed Products, shall not be accounted for in the calculation of sublicensing royalties.

 

4)                                            Licensee shall pay NIH an amendment issue royalty in the sum of five hundred thousand US Dollars ($500,000.00) as follows:

 

i)                                          Two hundred and fifty thousand Dollars ($250,000) shall be paid by Licensee within sixty (60) days of the effective date of this First Amendment.

 



 

ii)              The remaining amount of two hundred and fifty thousand Dollars ($250,000) shall be paid to NIH upon execution by Licensee of any new sublicensing or partnership agreement or on the first anniversary of this First Amendment, whichever occurs first.

 

5)                                            In the event any provision(s) of the Agreement is/are inconsistent with Attachment 1, such provision(s) is/are hereby amended to the extent required to avoid such inconsistency and to give effect to payment information in such Attachment 1.

 

6)                                            All terms and conditions of the Agreement not herein amended remain binding and in effect.

 

7)                                            The terms and conditions of this First Amendment shall, at NIH’ sole option, be considered by NIH to be withdrawn from Licensee’s consideration and the terms and conditions of this First Amendment , and the First Amendment itself, to be null and void, unless this First Amendment is executed by Licensee and a fully executed original is received by NIH within sixty (60) days from the date of NIH signature found at the Signature Page.

 

8)                                            This First Amendment is effective on                                                                                upon execution by all parties.

 



 

FIRST AMENDMENT TO L-116-2011/0

 

SIGNATURE PAGE

 

In Witness Whereof, the parties have executed this First Amendment on the dates set forth below. Any communication or notice to be given shall be forwarded to the respective addresses listed below.

 

For NIH :

 

 

 

 

 

/s/ Richard U. Rodriguez

 

5-23-13

Richard U. Rodriguez

 

Date

Director, Division of Technology Development and Transfer

 

 

Office of Technology Transfer

 

 

National Institutes of Health

 

 

 

Mailing Address or E-mail Address for Agreement notices and reports:

 

Chief, Monitoring & Enforcement Branch, DTDT
Office of Technology Transfer
National Institutes of Health
6011 Executive Boulevard, Suite 325
Rockville, Maryland 20852-3804 U.S.A.

 

E-mail:  LicenseNotices_Reports@mail.nih.gov

 

For Licensee (Upon information and belief, the undersigned expressly certifies or affirms that the contents of any statements of Licensee made or referred to in this document are truthful and accurate.):

 

/s/ John Alday

 

5-31-13

John Alday, CEO UniQurebiopharm B.V.

 

Date

 

I.  Official and Mailing Address for Agreement notices:
Chief Executive Officer:
Legal@uniqure.com

 

II.  For invoices, payments, and Financial notices (including royalty payments):
Finance Dept
Finance@uniqure.com

 

uniQure biopharma B.V.
Meibergdreef 61
1105BA Amsterdam
The Netherlands

 

Phone:                                                           0031 205667394

Fax:                                                                        0031 20 566 9272

 

Any false or misleading statements made, presented, or submitted to the Government, including any relevant omissions, under this Agreement and during the course of negotiation of this Agreement are subject to all applicable civil and criminal statutes including Federal statutes 31 U.S.C. §§3801-3812 (civil liability) and 18 U.S.C. §1001 (criminal liability including fme(s) or imprisonment).

 



 

ATTACHMENT 1 - ROYALTY PAYMENT OPTIONS

 

The OTT License Number MUST appear on payments, reports and correspondence.

 

Automated Clearing House (ACH) for payments through U.S. banks only

 

The NIH encourages our licensees to submit electronic funds transfer payments through the Automated Clearing House (ACH). Submit your ACH payment through the U.S. Treasury web site located at: https://www.pay.gov . Locate the “NIH Agency Form” through the Pay.gov “Agency List”.

 

Electronic Funds Wire Transfers

 

The following account information is provided for wire payments. In order to process payment via Electronic Funds Wire Transfer sender MUST supply the following information within the transmission:

 

Drawn on a U.S. bank account via FEDWIRE should be sent directly to the following account:

 

Beneficiary Account:

Federal Reserve Bank of New York or TREAS NYC

Bank:

Federal Reserve Bank of New York

ABA#

021030004

Account Number:

75080031

Bank Address:

33 Liberty Street, New York, NY 10045

Payment Details:

License Number (L-XXX-XXXX)

 

Name of Licensee

 

Drawn on a foreign bank account should be sent directly to the following account. Payment must be sent in U.S. Dollars (USD) using the following instructions:

 

Beneficiary Account:

Federal Reserve Bank of New York/ITS or FRBNY/ITS

Bank:

Citibank N.A. (New York)

SWIFT Code:

CITIUS33

Account Number:

36838868

Bank Address:

388 Greenwich Street, New York, NY 10013

Payment Details (Line 70):

NIH 75080031

 

License Number (L-XXX-XXXX)

 

Name of Licensee

Details of Charges (Line 71a):

Charge Our

 



 

Checks

 

All checks should be made payable to “NIH Patent Licensing”

 

Checks drawn on a U.S. bank account and sent by US Postal Service should be sent directly to the following address:

 

National Institutes of Health (NIH)
P.O. Box 979071
St. Louis, MO 63197-9000

 

Checks drawn on a U.S. bank account and sent by overnight or courier should be sent to the following address:

 

US Bank
Government Lockbox SL-MO-C2GL
1005 Convention Plaza
St. Louis, MO 63101
Phone: 314-418-4087

 

Checks drawn on a foreign bank account should be sent directly to the following address:

 

National Institutes of Health (NIH)
Office of Technology Transfer
Royalties Administration Unit
6011 Executive Boulevard
Suite 325, MSC 7660
Rockville, Maryland 20852

 



 

NATIONAL INSTITUTES OF HEALTH
SECOND AMENDMENT TO L-116-2011/0

 

This is the second amendment (“ Second Amendment ”) of the agreement by and between the National Institutes of Health (“ NIH ”) within the Department of Health and Human Services (“ HHS ”), and UniQure biopharma B.V. (formerly Amsterdam Molecular Therapeutics (AMT) B.V.) having an effective date of August 10, 2011 as amended for the first time on May, 31, 2013, and having NIH Reference Number L-116-2011/0  and L-116-2011/1 respectively (“ Agreement ”). This Second Amendment , having NIH Reference Number L-116-2011/2, is made between the NIH through the Office of Technology Transfer, NIH , having an address at 6011 Executive Boulevard, Suite 325, Rockville, Maryland 20852-3804, U.S.A., and UniQure biopharma B.V. (formerly Amsterdam Molecular Therapeutics (AMT) B.V.), having an office at Meibergdreef 61, 1105 BA Amsterdam, The Netherlands (“ Licensee ”). This Second Amendment includes, in addition to the amendments made below, a Signature Page.

 

WHEREAS, NIH and Licensee desire that the Agreement be amended a second time as set forth below in order to a) clarify the nonexclusive Field of Use , b) to update appendices D and E of the Agreement , and c) to update Article 6.13 of the Agreement with the name of an Exempt Collaborator that is approved to work with the Licensee on one Ultra-Orphan Indication.

 

NOW, THEREFORE, in consideration of the mutual covenants and promises contained herein, NIH and Licensee , intending to be bound, hereby mutually agree to the following:

 

1)              In Appendix B replace Paragraph I(b) of the Licensed Field of Use with the following:

 

(b)                                  Non-Exclusive Licensed Field of Use :  Use of the Licensed Patent Rights for the development and sale of AAV5 based therapeutic products to treat any human disease in any manner, where the treatment of such disease in such manner is not included in the Exclusive Licensed Field of Use .

 

2)              In Article 6.13 add the following:

 

(h)                                  Institut Pasteur has been approved by the NIH as an Exempt Collaborator for a clinical work related to Sanfilippo B.

 

3)              Replace Appendix D with Appendix D attached to this Second Amendment as EXHIBIT 1.

 

4)              Replace Appendix E with Appendix E attached to this Second Amendment as EXHIBIT 2.

 

5)              All terms and conditions of the Agreement not herein amended remain binding and in effect.

 

6)              The terms and conditions of this Second Amendment shall, at NIH sole option, be considered by NIH to be withdrawn from Licensee ’s consideration and the terms and conditions of this Second Amendment , and the Second Amendment itself, to be null and void, unless this Second Amendment is executed by Licensee and a fully executed original is received by NIH within sixty (60) days from the date of NIH signature found at the Signature Page.

 

7)              This Second Amendment is effective on the date of execution by the last party to execute this Second Amendment .

 



 

SECOND AMENDMENT TO L-116-2011/0

 

SIGNATURE PAGE

 

In Witness Whereof, the parties have executed this Second Amendment on the dates set forth below. Any communication or notice to be given shall be forwarded to the respective addresses listed below.

 

For NIH :

 

/s/ Richard U. Rodriguez

 

11-6-13

Richard U. Rodriguez

 

Date

Director, Division of Technology Development and Transfer

 

 

Office of Technology Transfer

 

 

National Institutes of Health

 

 

 

 

 

Mailing Address or E-mail Address for Agreement notices and reports:

 

 

 

Chief, Monitoring & Enforcement Branch, DTDT

 

 

Office of Technology Transfer

 

 

National Institutes of Health

 

 

6011 Executive Boulevard, Suite 325

 

 

Rockville, Maryland 20852-3804 U.S.A.

 

 

 

 

 

E-mail: LicenseNotices_Reports@mail.nih.gov

 

 

 

For Licensee (Upon information and belief, the undersigned expressly certifies or affirms that the contents of any statements of Licensee made or referred to in this document are truthful and accurate.):

 

/s/ Piers J. Morgan P.J. Morgan

 

November 11, 2013

Piers J Morgan, CFO, uniQure biopharma B.V.

 

Date

I

Official and Mailing Address for Agreement notices:

 

 

 

Chief Executive Officer;

 

 

 

Legal@uniqure.com

 

 

II

For invoices, payments, and Financial notices (including royalty payments):

 

 

 

Finance Dept

 

 

 

Finance@uniqure.com

 

 

 

 

uniQure biopharma B.V.

Meibergdreef 61

 

 

1105BA Amseterdam

 

 

The Netherlands

 

 

 

 

 

Phone: 0031 205667394

 

 

 

 

 

Fax: 0031 20 566 9272

 

 

 

Any false or misleading statements made, presented, or submitted to the Government , including any relevant omissions, under this Agreement and during the course of negotiation of this Agreement are subject to all applicable civil and criminal statutes including Federal statutes 31 U.S.C. §§3801-3812 (civil liability) and 18 U.S.C. §1001 (criminal liability including fine(s) or imprisonment).

 



 

Exhibit 1

 

APPENDIX D — BENCHMARKS AND PERFORMANCE (L-116/2011)

 

Licensee agrees to the following Benchmarks for its performance under this Agreement and, within thirty (30) days after achieving a Benchmark , shall notify PHS that the Benchmark has been achieved.

 

Note:                   No formal Phase III clinical trial is required for Marketing Approval for any Orphan Indication

 

Benchmarks for a Licensed Product of Orphan Indication - liver

 

I.                                         Initiation of first Phase I clinical trial or foreign equivalent — 2012

 

II.                                    Initiation of first Phase II clinical trial or foreign equivalent — 2016

 

III.                               Submission to Regulatory Authority of first Marketing Approval or foreign equivalent — 2018

 

Benchmarks for a Licensed Product - brain

 

I.                                         Initiation of Preclinical Development phase or foreign equivalent — 2011

 

II.                                    Initiation of first Phase I clinical trial or foreign equivalent — 2013

 

III.                               Initiation of first Phase II clinical trial or foreign equivalent — 2016

 

IV.                                Submission to Regulatory Authority of first Marketing Approval or foreign equivalent —2018

 



 

Exhibit 2

 

APPENDIX E - COMMERCIAL DEVELOPMENT PLAN (L-116-2011)

 

The table below (table 1) presents a comprehensive list of all uniQure research and development projects utilizing the Licensed Patent Rights , according to main disease site and divided into projects that are, a) commercial projects, b) already in development stages, c) active research (there is already internal research activity ongoing and d) exploratory research projects (currently being considered as potential projects worth further investigation in the near future).

 

Table 1: uniQure R&D projects

 

 

 

Liver (AAV5 based)

 

Brain & CNS (AAV5 based)

Commercial
Projects

 

 

Development Projects

 

·                   Acute Intermittent Porphyria (AMT-021)

·                   Hemophilia B (AMT-060)

 

·                   Sanfilippo B (MPS IIIB)
(AMT-110)

Active Research Projects

 

·                   Hemophilia A (HA)

·                   Cirrhosis

·                   Hyperoxalurea (HP1)

 

·                   Huntington’s Disease

·                   Multiple System Atrophy (MSA)

·                   Hearing loss

Exploratory Research Projects

 

·                   Phenylketonuria (PH1)

·                   Ornithine transcarbamylase deficiency (OTCD)

·                   Glycogen storage disease type II (Pompe)

·                   Maroteaux-Lamy syndrome (MPS VI)

·                   Fabry disease

 

·                   Amyotrophic lateral sclerosis (ALS - motor neurone disease or Lou Gehrig’s disease)

·                   Spinal muscular atrophy (SMA)

·                   Batten’s disease

·                   Tay Sachs

·                   Krabbe disease (globoid cell leukodystrophy or galactosylceramide lipidosis)

 

Detailed information on the development and active research projects is provided below.
NOTE:  All dates contained in this Commercial Development Plan are projected estimates only.

 



 

Liver Programs

 

A) Development Programs

 

1.               AMT-021 for Acute Intermittent Porphyria

 

1.1.1                           Disease Background

 

Acute Intermittent Porphyria, or AIP, is a rare liver metabolic disorder resulting from mutations in the PBGD gene. This gene encodes for the enzyme porphobilinogen deaminase (also known as hydroxymethylbilane synthase — HMBS), a liver protein necessary for the production of heme, a component of hemoglobin and other blood proteins. Insufficient activity of this protein leads to an accumulation of toxic metabolites (ALA and PBG), resulting in a wide variety of serious clinical problems, including acute, severe abdominal pain, muscular weakness and an array of neurologic manifestations, including psychiatric episodes, seizures and coma. In the majority of cases, attacks are triggered by precipitating factors such as hormonal fluctuations, infections, drugs and dietary changes. Long-term consequences may include irreversible nerve damage, liver cancer and kidney failure. Patients with AIP experience regular hospitalizations and extremely poor quality of life, and may in some cases require liver transplants. Acute attacks can be life-threatening. Current therapies only target the disease symptoms and do not prevent attacks or fully minimize or control their consequences.

 

A recent epidemiological study reported that, in Europe (excluding Sweden), the incidence of AIP is 0.13 per million population per year and based on that they estimated a prevalence of 5.9 per million population (Elder et al., 2012). In Sweden the incidence and prevalence of AIP are about four times higher than in the rest of Europe due to a founder effect originating in Lappland (Floderus et al., 2002).  The frequency in the United States is estimated to be 1-5 cases per 100,000 population ( www.emedicine.medscape.com/article/205220-overview#a0199 ).

 

1.1.2                           Overview of AMT-021 Program

 

The goal of our AMT-021 program is to provide long-term normalization of the PBGD protein in order to prevent acute AIP attacks and their complications.

 

The program has been developed through a collaborative agreement with the Foundation for Applied Medical Research (FIMA), its Center for Applied Medical Research (CIMA) and its commercialization arm, DIGNA Biotech, of the University of Navarra (Pamplona, Spain). Part of the funding to support for the Phase I trial (including GLP safety & toxicology studies and the observational trial) was secured through the European Commission Framework Programme 7  award (€3.3 million, grant agreement 261506) made to the AIPGENE consortium ( www.aipgene.org/ ), of which uniQure is a partner.

 

UniQure holds an exclusive license to the gene cassette being used in the Phase I clinical trial. Under our agreement with DIGNA Biotech and the other consortium members, Licensee have an exclusive right to all data related to the program.

 

1.1.3                           Preclinical Development

 

·                   Product Profile

 

1.1.4                           AMT-021 is designed to be delivered systemically through a peripheral vein in a single administration.

 

AMT-021 or rAAV5-hPBGD, is a recombinant adeno-associated vector of serotype 5, consisting of:

 

·                   Inverted terminal regions or ITRs of the adeno-associated serotype 2

 

·                   A human codon optimized porphobilinogen deaminase gene or hPBGDco as the therapeutic gene

 

·                   A liver specific promoter constituted by the albumin enhancer (Ealb) and the alfa-1-antitrypsin promoter (hAAT)

 



 

1.1.5

 

·                   Pre-clinical Proof of Concept

 

1.1.6                           Pre-clinical proof of concept (PoC) studies have been performed using the AIP murine model developed by Lindberg et al. (1999).  In these studies, long term therapeutic efficacy was achieved.  More specifically, at 5x10 13  gc/kg, metabolic correction of the hepatic PBGD enzyme activity, normalization of the PBG and ALA precursor’s accumulation in urine and improvement of the motor coordination were observed. Additionally, a complete neurological study indicated the correction of neurotoxic porphyrin precursors was able to restore nerve conduction and the impaired peripheral neuropathy.

 

In non-human primates (NHP) treated with AMT-021 at a dose of 5x10 13  gc/kg endogenous PBGD enzymatic activity increased by a factor of two in male and between three and five times in female animals.

 

·                   Non-clinical safety & toxicology studies

 

1.1.7                           The following table presents a summary of the AMT-021 non-clinical safety and toxicology studies that have been conducted to support the clinical development program.

 

Parameter to be
assessed

 

Study performed

 

Results

Pharmacokinetics

 

1) GLP biodistribution in C57Bl/6 mice (180 days) with validated QPCR method.

 

2) Biodistribution data in Rhesus macaques (supportive)

 

In both species,

·                   Clear correlation between the dose of vector administered and the genome copies of the vector as well as vector RNA (expression) quantified in the liver.

·                   Tissue biodistribution was mainly limited to liver although some significant transduction was detected in spleen, lymph nodes, heart and adrenal glands.

Toxicity

 

1) GLP toxicity study in Rhesus macaques, 30 days

 

2) GLP toxicity study in C57Bl/6 mice (180 days)

 

In Rhesus macaques:

·                   No specific macroscopic or microscopic findings, no severe immune/inflammatory reactions (NOAEL 5x10 13  gc/kg)

In C57Bl/6 mice:

·                   No severe adverse toxicologically significant findings

·                   At highest dose limited hepatocyte vacuolation due to metabolic adaptation to high PBGD activity in liver (NOAEL 5x10 14  gc/kg)

 



 

Off-target expression

 

Included in the GLP toxicity study in monkey and mouse

 

Vector RNA analysis for all tissues other than liver showed that there is no PBGD transcription and translation elsewhere.

Shedding pattern

 

Included in the Rhesus macaques study

 

Shedding in semen and saliva is transient (cleared by day 30)

Persistence in semen and risk of germline transmission

 

Mouse Germline transmission study in C57Bl/6 mice

 

No transmission to offspring

Carcinogenicity

 

Analyze tissues from the Rhesus studies using LAM-PCR

 

·                   Low frequency of integration sites, with random distribution

·                   No preferences for CpG islands

·                   No preference for palindromic regions

·                   No hotspots (low clustering level)

 

1.1.8                                                                                              Summary of AMT-021 Preclinical Development Program

 

Single intravenous administration of AMT-021 into wild type mice and Rhesus macaques results in:

 

·                   Efficient liver transduction resulting in dose dependent increase in viral RNA copy numbers and in turn producing increased PBGD activity

 

·                   No morbidity, no changes in body weight or food intake

 

·                   No changes in biochemistry, hematology, coagulation and urinalysis associated with AAV5-hPBGD

 

·                   Negative vector shedding 30 days after viral administration in serum, saliva, nasal secretions, urine, faeces and semen

 

·                   Tissue biodistribution that is mainly limited to liver although some significant transduction was detected in spleen, lymph nodes, heart and adrenal glands

 

·                   Specific hepatic PBGD expression

 

1.1.9                           Clinical Development Program

 

1.1.10                    The key regulatory and clinical development best estimate milestones for AMT-021 include the following,

 



 

·

 

EMA Orphan Drug Designation (EU/3/09/632)

 

Apr 2009

 

 

 

 

 

·

 

FIMA/ CITA/ UTE/ DIGNA - AMT Collaborative Agreement

 

 

May 2010

 

 

 

 

 

·

 

EU-FP7 AIPGene Consortium

 

Jan 2011

 

 

 

 

 

·

 

Observational Study AEMPS approval

 

May 2011

 

 

 

 

 

·

 

Observational Study start

 

Jul 2011

 

 

 

 

 

·

 

Phase I Study AEMPS approval

 

Oct 2012

 

 

 

 

 

·

 

Phase I Study: first patient treated

 

Dec 2012

 

 

 

 

 

·

 

Phase I Study: last patient treated

 

Aug 2013

 

Expected milestones

 

·

 

Phase II/III start:

 

 

 

 

4Q2016

·

 

MAA/ NDA submission:

 

 

 

 

 

·

 

Observational trial

 

2Q2018

 

1.1.11                    A prospective non-interventional (pre-treatment) observational study started at the end of 2011 that aims to assess the evolution of disease-related clinical and laboratory parameters in time, as well as characterize aspects of disease management such as AIP-related hospitalization. This baseline assessment is intended to study possible relationships between biochemical parameters and clinical endpoints that will in turn be valuable in evaluating any signs of efficacy in the Phase I trial as well as in subsequent trials. Eight patients are expected to be enrolled who after completion of this observational phase would then enter the interventional stage of the program, i.e., first-in-human clinical study (Phase I). The observational study is to last for at least six months for each participant.

 

1.1.12                    To date all 8 AIP-patients have been recruited into the observational study and all but one have completed a minimum of 6 months pre-treatment assessments.  The last patient completed the observational study in August 2013.

 

·                   Phase I trial

 

1.1.13                    The Investigational Medicinal Product Dossier (IMPD) was submitted to the AEMPS (Spanish Agency for Medicines and Medical Devices) in June 2012 and was approved by the Agency in October 2012.

 

The Phase I study is a multicenter, open label, prospective, interventional, single dose, dose-escalation clinical trial to investigate the safety and tolerability of AAV5-hPBGDco (AMT-021) in patients with severe Acute Intermitted Prophyria (Eudra CT no. 2011-005590-23).

 

The primary objective is to assess the safety of systemic administration and determine the maximum tolerated doses (MTD). Secondary objectives include urinary levels of toxic metabolites (ALA and PBG), disease symptoms evaluation, quality of life evaluation and assessment of pharmacokinetics. Exploratory objectives include, neurological involvement, identification of novel biomarkers and pharmacokinetic modeling.

 



 

The Phase I study was initiated in December 2012 in the Department of Medicine (Liver Unit) at the University Clinic of the University of Navarra (Pamplona, Spain). There are two patients per cohort and four cohorts in the trial (each cohort receiving 5x10 11 , 2x10 12 , 6x10 12  or 1.8x10 13  gc/kg) and all patients will be followed- up for one year as part of the Phase I study.

 

All 8 patients who completed the observational trial have also been treated as part of the Phase I study.  In the 8 treated patients, no Serious Adverse Events, Treatment Emergent Adverse Events or Liver Events (Dose Limiting Toxicities - DLT’s) related to the study medication have been observed to date.

 

·                   Future Clinical Development

 

It is envisaged that the Phase II/III will be a confirmatory trial where the study population and the outcomes to be assessed (efficacy endpoints — clinical and biochemical) will be based on those as for Phase I. Licensee also intend to carry out the study in both Europe and the USA.

 

1.1.14                                                                                       Summary of AMT-021 Clinical Development Program

 

·                   The first time an AAV5 gene therapy product has been tested in humans

 

·                   The first time an AAV gene therapy product has been tested in humans at such high dose, i.e., 1.8x10 13  gc/kg

 

·                   No Serious Adverse Events, Treatment Emergent Adverse Events or Liver Events (DLT’s) related to the study medication have been observed in the Phase I study to date

 

·                   The Phase I is expected to be completed in 3Q14 and Phase II/III is expected to start by the end of 2014

 

·                   The Phase II/III program will run in parallel in Europe and US where MAA and NDA, respectively, are expected in 4Q2016

 

2.               AMT-060 for Hemophilia B

 

1.1.1                           Disease Background

 

Hemophilia B is a serious inherited orphan disease in males characterized by insufficient blood clotting. The condition can lead to repeated and sometimes life-threatening episodes of external and internal bleeding following accidental trauma or medical interventions. The episodes may cause long-term damage, for example to the joints, and may be fatal if they occur in the brain. The deficient blood clotting is caused by the lack of functional human Factor IX, or hFIX, a blood clotting factor, as a result of mutations in the gene responsible for encoding this essential protein. The presence of hFIX at greater than 1% of normal levels has a therapeutic effect in promoting clotting. The current standard treatment is prophylactic protein replacement therapy, in which frequent intravenous administrations of recombinant Factor IX (often 2-3 times per week) are

 



 

required to stop or prevent bleeding. Protein replacement therapy is costly ($150,000-200,000 per patient per year) and burdensome, and does not completely prevent bleeding.

 

The total Hemophilia B patient population in the European Union and the United States is estimated at approximately 25,000, according to the World Federation of Hemophilia 2010 Report on the Annual Global Survey. About 40% of individuals with the disease have a severe disorder, characterized by functional factor IX levels that are less than 1% of normal, whereas moderately severe Hemophiliacs (about 30% of the Hemophiliac population) have 1%-5% of normal and those with the mild phenotype (the remaining 30%) have between 5% and 40% of normal factor IX levels ( www.orpha.net). Based on these estimates Licensee believes that approximately 70-85% of the worldwide patient population would be eligible for treatment with gene therapy. Licensee believes that the treatment would not be appropriate for those patients with very mild disease phenotype.

 

1.1.2                           Overview of AMT-060 Program

 

The goal of our AMT-060 program is to restore blood clotting on a long-term basis through the introduction of the functional gene for hFIX into the patient’s liver cells.  Licensee is currently in the process of finalizing pivotal (GLP) safety and toxicology studies and preparing to conduct a Phase I trial.

 

1.1.3                           Preclinical Development

 

·                   Product Profile

 

1.1.4                           AMT-060 is designed to be delivered systemically through a peripheral vein in a single administration.

 

The use of recombinant adeno-associate vectors (rAAV) of serotype 5 (rAAV5) for targeted gene delivery to the liver was pioneered by St. Jude Children’s Research Hospital (SJCRH) where for pre-clinical experiments the hFIX expression cassette was packaged into AAV5 capsids in HEK-293T mammalian cells. HEK-293 produced AAV5-hFIX is not suitable for further development because as a production system it is not amenable to large-scale production. To allow up scaling, the expression cassette has now been transferred into uniQure’s proprietary baculovirus expression vector system (BEVS) that can be adapted to a GMP setting. The resulting vector produced using the baculovirus expression system is termed AAV5-hFIXco or AMT-060.  Licensee also holds a license from SJCRH to the gene cassette used in the currently ongoing Phase I/II AAV 2/8-LP1-hFIXco trial.

 

AMT-060, rAAV5-hFIXco, is a recombinant adeno-associated vector of serotype 5, consisting of:

 

·                   Inverted terminal regions (or ITRs) of the adeno-associated serotype 2

 

·                   A human codon optimized FIX gene (or hFIXco) as the therapeutic gene

 

·                   The liver specific promoter, LP1, derived from the human apolipoprotein hepatic control region and the human alpha-1-antitrypsin (or hAAT) promoter

 



 

·                   Virus serotype selection

 

The hFIXco expression cassette and rAAV5 or AAV8 vectors have been extensively studied in mice and non-human primate.  Both vectors have been shown to have similar tropism to (preference to transduce) the liver (Nathwani et al., 2007) and AAV5-hFIXco was shown to mediate expression of significant levels of human factor IX in non-human primates (NHP) during a monitoring period of more than 5 years (Nathwani et al., 2011). In this study none of the animals presented elevated liver enzymes levels or other signs of toxicity during the whole observation period. Liver examination by MRI scanning did not reveal any abnormalities in any of the animals.

 

These pre-clinical data suggest that i.v. administration of the AAV5-hFIXco vector is able to mediate a similar level of human factor IX as presented for AAV8-hFIXco, and such administration is not associated with safety concerns or immunogenicity against the human factor IX.

 

·                   Pre-clinical Proof of Concept

 

Pre-clinical PoC studies have been carried out in wild type mice, non-human primates (NHP) and are currently being completed in transgenic Hemophilia B mice. In wild type mice (C57Bl/6) intravenous administration of AMT-060 mice resulted in dose-dependent levels of (human) factor IX levels in murine plasma as determined by ELISA. Human factor IX levels amounted up to 11% of those in normal human plasma 4 weeks after infusion of 5x10 12  gc/kg, demonstrating that AAV5-hFIXco produced in the BEVS is biologically active.

 

In Rhesus monkeys dosed with AMT-060 (5x10 12  gc/kg) by intravenous infusion, human FIX levels peaked to 7%-16% of normal human levels one week after infusion, and stabilized to 5-10% of normal human levels 4 weeks after infusion until sacrifice (12 weeks after dosing). These kinetics are in accordance with those observed in previous studies (Nathwani et al., 2007; Jiang et al., 2006), indicating that i.v. administration of AAV5-hFIXco produced in BEVS results in a level of factor IX in plasma that is similar to that produced using AAV5-hFIXco produced in HEK293 cells. Post mortem, (RT)-QPCR demonstrated homogeneous vector DNA delivery and transgene expression in the liver. No signs of adverse reactions were observed. Infusion was associated with slight and transient effects in plasma chemistry shortly after dosing, such as a brief increase of liver enzyme activity levels, consistent with infusion of a viral protein. Necropsy revealed no significant macroscopic or microscopic abnormalities.

 

Preliminary data in Hemophilia B mice indicate that treatment with AMT-060 induces normalization of FIX levels as well as clotting time.

 

·                   Non-clinical safety & toxicology studies

 

The following table presents a summary of the AMT-060 non-clinical safety and toxicology studies that are being conducted to support the clinical development program.

 



 

Parameter to be
assessed

 

Study performed

 

Status

Pharmacokinetics

 

1) GLP biodistribution in C57Bl/6 mice (180 days) with validated QPCR method.
2) Biodistribution data in Rhesus macaques (supportive)

 

Ongoing

Toxicity

 

1) GLP toxicity study in Cynomolgus monkeys (180 days)
2) GLP toxicity study in C57Bl/6 mice (180 days)
3) Liver pathology in Rhesus macaques (supportive)

 

Ongoing

Off-target expression

 

Included in the GLP toxicity study in monkey and mouse

 

Ongoing

Shedding pattern

 

Partly included in the mouse GLP toxicity study.
Partly included in the Rhesus macaques study.

 

Ongoing

Persistence in semen and risk of germline transmission

 

1) Mouse Germline transmission study in C57Bl/6 mice
2) QPCR of the testis in the Rhesus macaques at 90 days (non-GLP).

 

Ongoing

Carcinogenicity

 

Analyze tissues from the Rhesus studies using LAM-PCR

 

Ongoing

Juvenile toxicity

 

Ethical approval obtained and study is being planned (to support PIP plans)

 

Ongoing

 

1.1.5                                                                                              Summary of AMT-060 Preclinical Development Program

 

·                   AAV5-hFIXco shows similar liver tropism to AAV8-hFIXco and results in significant and long lasting increase in FIX expression.

 

·                   Single intravenous administration of AMT-060 into wild type mice and Rhesus macaques results in significant and long lasting hFIX levels with no noticeable adverse events and no macroscopic or microscopic findings.

 

·                   GLP safety and toxicology studies are expected to be completed in December 2013.

 

1.1.6                           Clinical Development Program

 

1.1.7                           The key regulatory and clinical development milestones for AMT-060 include the following:

 

·

 

EMA Orphan Drug Designation:

 

Nov 2011

 



 

·

 

FDA Orphan Drug Designation:

 

Jan 2012

 

 

 

 

 

·

 

EMA Scientific Advice:

 

Feb 2012

 

 

 

 

 

·

 

EMA Phase I Protocol Advice:

 

Aug 2012

 

 

 

 

 

·

 

GLP Safety & Tox Studies:

 

2Q2012 to 4Q2013

 

Expected milestones

 

·

 

IMPD submission:

 

2Q2014

 

 

 

 

 

·

 

Phase I start:

 

3Q2014

 

 

 

·

 

Phase II/III start:

3Q2016

 

 

 

·

 

MAA/ NDA submission:

 

2Q2018

 

 

 

 

 

·

 

Phase I trial

 

 

 

The Phase I study will be a multicenter, open label, prospective, interventional, single dose, dose-escalation clinical trial to investigate the safety and tolerability of AAV5-hFIXco (AMT-060) in patients with severe Hemophilia B.

 

The primary objective is to assess the safety of systemic administration and determine the maximum tolerated doses (MTD). Secondary objectives include:

 

·                   To estimate the appropriate dose required to achieve stable expression of hFIX at or above 3% of normal

·                   To evaluate kinetics (dose-related duration and magnitude) of expression

·                   To assess the immune response to hFIX transgene product

·                   To assess the immune response to the AAV5 capsid proteins

·                   To assess viral shedding in various body fluids (including semen)

·                   To assess the occurrence of FIX inhibitors

·                   To evaluate coagulation parameters

·                   To assess need for FIX concomitant treatment

 

Twelve male adults patients ( > 18 year old to < 35 year old) with genetically confirmed Hemophilia B and phenotypically defined as having severe disease ( < 1% of normal plasma FIX levels) are expected to be enrolled.  Initial patient follow-up will last for 12 months as part of the Phase I trial.

 

·                   Future Clinical Development

 

1.1.8                           It is envisaged that the Phase II/III will be a confirmatory trial where the study population and the outcomes (efficacy endpoints — clinical and biochemical) will be based on those for the Phase I. Licensee will also consider expanding the patient population to moderately severe patients and intend to carry out the study in both Europe and USA.

 

1.1.9                                                                                              Summary of AMT-060 Clinical Development Program

 

·                   The IMPD is planned to be submitted in 2Q2014

 



 

·                   Phase I is planned in patients with severe Hemophilia B and is expected to start in 3Q2014

 

·                   It is the intention that in Phase II/III the patient population will expand to moderately severe Hemophilia B patients

 

·                   The Phase II/III program will run in parallel in Europe and USA where MAA and NDA, respectively, are expected in 2Q2017

 

The Hemophilia B program has been partnered with Chiesi. The co-development agreement has ben shared with NIH .

 

B) Active Research Projects

 

1.               Hemophilia A

 

Disease Background : Hemophilia A (HA) is a genetic, X-linked, recessive disorder caused by production of dysfunctional or by production of insufficient amount of factor VIII (FVIII) protein, a key protein involved in the blood coagulation cascade.  Hemophilia A patients suffer from spontaneous bleeding in the large joints and soft tissue, and are at risk for intracranial hemorrhage.  Recurrent episodes of joint bleeding can lead to crippling arthropathy, particularly in severely affected patients.  HA comprises the majority of hemophilia patients (80%), with incidence of ~1:10,000 to 1:50,000 males affecting 400,000 people worldwide.

 

Numerous mutations in the FVIII gene have been described giving rise to different disease phenotypes.  Similarly to Hemophilia B (HB), individuals with less than 1% active factor are classified as having severe hemophilia, those with 1—5% active factor have moderate hemophilia, and those with mild hemophilia have between 5—40% of normal levels of active clotting factor.

 

Clinical need : HA seems an excellent candidate for gene therapy (GT) as it is a well characterized monogenic disorder.  The product of the FVIII gene is a plasma protein which is normally secreted by hepatocytes and endothelial cells but can also be expressed in other cell types, e.g., adipocytes, mycoytes or fibroblasts.  Furthermore, only modest increase >1% can markedly reduce spontaneous bleedings.  The effects of gene therapy can be readily monitored by changes in phenotype and by obtaining peripheral blood to measure FVIII antigen levels and clotting factor activity.  Currently, treatment for HA consists of infusion of either plasma-derived or rFVIII protein for bleeding episodes.  Although, prophylactic infusion of FVIII concentrates is generally effective in alleviating bleeding episodes and subsequent joint disease, the short half-life of FVIII (~12 hours) and the high cost of purified FVIII products make life-long prophylactic treatment demanding for patients and costly.

 

Feasibility

 

Gene : The gene of factor VIII is located on the long arm of the X chromosome.  It spans over 180 kb, and as such is one of the largest genes known.  It comprises of 26 exons, which encode a polypeptide chain of 2351 amino acids including a signal peptide of 19 and a mature protein of 2332 amino acids.  It is a secreted protein.  Its primary structure, deduced from the cloned factor VIII cDNA, includes discrete domain structure: A1-a1-A2-a2-B-a3-A3-C1-C26-8.  The B domain is unique in that it exhibits no significant homology with any other known protein and can be deleted with the resulting recombinant protein displaying essentially normal survival in circulation and able to correct the bleeding tendency in HA patients.

 

Vector optimization : A rAAV5 containing HLP-hFVIIIco (promoter HLP; codon optimized hFVIII with partial B-domain deletion) has been generated in-house where the preserved B-domain consists of 225 N-terminal amino acids containing 6N-glycosilation sites. This variant has been previously shown to increase secretion of FVIII as

 



 

compared to wild type or to FVIII with complete B-domain deletion and in-house work with HLP-hFVIII has shown that it produces active FVIII protein in vitro and in vivo .

 

This FVIII expression cassette size is however ~5.6 kbp, which exceeds the AAV packaging limit (4,7-4,9 kbp).  In-house molecular analysis studies of the encapsidation products showed that the 5.6kbp FVIII expression cassette is not entirely encapsidated in the AAV particle. Instead + and – DNA strands of the encapsidated molecules revealed missing 5’ ends. This is consistent with previously reported unidirectional (starting at 3’ end) packaging mechanism operating according to “head-full principia” with 4.7-4,9 kbp limit. Licensee hypothesizes that the correct template for production of FVIII was assembled in the target cell based on complementation of + and – DNA strains each of which delivering the 5’ missing end. This is to be expected since + and – strain complementation is responsible for normal rAAV episome formation.  Licensee proposes therefore to develop a product that consist of incomplete + and – DNA strains, which are assembled into complete expression cassette upon episome formation.

 

A proof of concept study has been initiated involving a number of FVIII construct and including full FVIII codon optimized gene.  The study aims to characterize the viral DNA, formation of episomes upon delivery of the expression cassette to the nucleus, resulting mRNA and FVIII protein.  The potency of the vector is currently being investigated in a number of animal models.

 

It is our aim to develop this product to clinical stage Phase I by the end of 2015. Duration of clinical development and further timelines have not been defined.

 

Development overview to IMPD:

 

 

Completion of vector optimization work will provide the first milestone (Go/No Go) for the project.

 

Safety Assessment : The disease and gene therapy approach are similar (or equivalent) to Hemophilia B where no major safety concerns have been described.

 

2.               Cirrhosis

 

Disease Background : Liver cirrhosis is a consequence of chronic liver disease characterized by replacement of liver tissue by fibrosis, scar tissue and regenerative nodules (resulting from regeneration of damaged tissue), leading to loss of liver function.  The four leading causes of cirrhosis and primary liver cancer in Europe include harmful alcohol consumption, viral hepatitis B, viral hepatitis C and metabolic syndromes related to overweight and obesity.  The European Association for the Study of the Liver in its 2013 report reported that approximately 29 million people in the European Union suffer from a chronic liver condition and that the incidence and prevalence of two conditions, cirrhosis and primary liver cancer, are key to understanding the burden of liver disease.  Both conditions represent the end-stage of liver pathology and thus are indicative of the associated mortality.

 



 

The hypothesis behind this project is that liver cirrhosis is a state of IGF-I insufficiency and low expression of IGF-I locally in the liver will revert and/ or prevent further exacerbation of cirrhosis.  A confidentiality agreement concerning this project was signed between DIGNA/ CIMA and uniQure in October 2012.

 

Pre-clinical evidence to support this hypothesis includes the following:

 

·                   Proof of Mechanism: SV40-IGF-I administration in rat cirrhotic liver models resulted in increased mRNA and protein levels of IGF and IGF-BP3 compared to control animals in the hepatocytes and levels remained constant for up to 6 months (Sobrevals et al., 2010).

 

·                   Proof of Principle: SV40-IGF-I mediated expression in rat cirrhotic liver models, induced fibrolysis through upreguation of MMPs and downregulation of TIMP-1/-2, reduced expression of pro-fiborgenic factors (TGFb, amphilregulin, PDGF, CTGF, VEGF), induced antifibrogenic and cytoprotective factors (HGF), promoted hepatocyte differentiation through upregulation of HNF4a and downeregulation of WT-1 (Sobrevals et al., 2010).

 

Treatment with recombinant IGF-I protein resulted in improved hepatic function, decreased oxidative stress/ damage, improved mitochondrial function and decreased fibrosis (Castillo-Cortazar et al., 2000; García-Fernández et al., 2005; Lorenzo-Zuniga et al., 2006).

 

·                   Proof of Concept: SV40-IGF-I-mediated expression in rat cirrhotic liver reversed fibrosis by decreasing expression of collagen I & IV and deactivation of HSC, and improved liver function through normalisation of AST, ALT, ALP, bilirubin and albumin (Sobrevals et al., 2010).

 

Clinical evidence to support disease linkage includes the following:

 

·                   In patients suffering from liver cirrhosis circulating IGF-I levels (or IGF-BP3) correlate with disease severity scores; Child-Pugh and MELD (Kratzsch et al., 2005; Khoshnood et al., 2013).

 

·                   A short course (for 4 months) of IGF-I recombinant therapy treatment increased the levels of albumin and tended to improve energy metabolism (surrogates for liver function) & the levels of serum albumin positively correlated with IGF-I/IGF-I BP3 ratio (Conchillo et al., 2005).

 

Clinical need : Transplantation is the only curative option for the disease and contraindications to transplantation include, a) co-morbidities (e.g., TB), b) over 65 years of age, c) coronary artery disease and d) tumours in previous 5 years.

 

The initial target population for IGF-I gene therapy for liver cirrhosis could/ would be those cirrhotic patients with IGF-I insufficiency (i.e., 50% of all cirrhotic patients), possibly patients with Child-Pugh A and/ or B score and with IGF-I levels below normal values.  An ODD application for this specific population may be considered.  The table below indicates the Child-Pugh scoring scheme for liver disease prognosis.

 

Points

 

Class

 

One Year
Survival

 

Two Year
Survial

 

5-6

 

A

 

100

%

85

%

7-9

 

B

 

81

%

57

%

10-15

 

C

 

45

%

35

%

 

Feasibility :

 

Gene: The IGF1 gene is located on chromosome 12 and spans 7.3 kb encoding a 70 amino acid residue protein. It contains 6 exons, 4 of which are alternatively spliced depending on tissue type and hormonal environment.  The IGF1 coding region is flanked by sequences encoding an amino-terminal peptide of at least 25 residues and a

 



 

carboxyl-terminal peptide of 35 amino acids which indicates that IGF1 is synthesized as a precursor protein that undergoes proteolytic processing at both ends before being secreted.

 

Vector optimization: The IGF-1 rat version is available with CIMA and was used for proof of concept studies.  Human IGF-I vectors are being developed in-house as part of work done by the Emerging Technologies Research Group on regulated gene expression systems.

 

Animal models: A rat model is available with CIMA and has been used for proof of concept studies.  A number of other small animal models have been described (Liu et al., 2013).

 

Biomarkers: Circulating IGF-I (and other related proteins) can be monitored using commercially available methodology.  However the relevance of this to liver (local) levels of IGF-I and whether GT can deliver sufficient amounts of IGF-I that that can be readily detectable in the circulation need to be established.

 

Liver function and signs of cirrhosis can be monitored following well established standard procedures (e.g., liver enzymes, markers of fibrosis etc.).

 

The PoC obtained at CIMA will have to be repeated with uniQure’s AAV5-IGF1 vector. Licensee is at the initial stages of research aiming to initiate a Phase I clinical trial by the end of 2016.

 

Development overview to IMPD:

 

 

The GLP safety and toxicology studies will provide the first milestone (Go/No Go) for the project.

 

Safety Assessment : Safety studies in rat disease models (8 months) and wild type rats  (8 weeks) showed no signs of toxicity due to treatment with SV40-IGF-I (Sobrevals et al., 2010).

 

Potential toxicity concerns include tumor formation and interference with insulin/ glucose metabolism albeit both issues are unlikely as the aim of this approach would be to upregulate levels of IGF-I where they are already below normal rather than to achieve supra-physiological levels.  In addition, gene therapy vectors are likely to induce lower level of localized expression without substantial increase in serum IGF-I levels.  Regarding potential for tumorigenesis, IGF-I therapy is thought to favor hepatocellular differentiation, i.e., opposes carcinogenesis, and studies have shown that sharp decrease in IGF-I in cirrhotic liver may contribute to hepatocellular carcinoma (HCC).  In addition it is believed that it is IGF-II that is the key player in HCC.  Furthermore, patients with existing tumor nodules in their liver could/ should be excluded from trials.

 

[NOTE: Hepatocellular carcinoma occurs at a rate of 1% to 4% per year after cirrhosis is established and cirrhosis underlies HCC in approximately 80%-90% of cases worldwide (Giovanna Fattovich  et al., 2004), i.e., the vast majority of cirrhotic patients do not develop HCC or at least they do not live live long enough to develop it]

 



 

3.               Hyperoxaluria

 

Disease Background : Primary hyperoxaluria type I (PH1) is a rare, autosomal recessive inherited metabolic disorder characterized by a deficiency of the hepatic enzyme alanine-glyoxylate aminotransferase (AGXT), which produces a marked increase in endogenous oxalate synthesis by the liver. Oxalate is a metabolic end product in humans and excess oxalate provokes hyperoxaluria, causing progressive urolithiasis, nephrocalcinosis and chronic renal failure, ultimately leading to end-stage renal failure (ESRF) and death if untreated.

 

It is the most common and severe variant among a spectrum of metabolic disorders resulting in hyperoxaluria.  The disease has an estimated prevalence ranging from 1 to 3 per 1 million individuals and an estimated incidence of 1-9:100,000 live births per year in Europe. However, higher rates are reported in historically isolated populations, like the Canary Islands. PH1 accounts for <1% of pediatric ESRF in developed countries.

 

A pre-clinical proof of concept study has already been conducted in collaboration with Eduardo Salido (University Hospital of Canary Islands) using AGXT knockout mice demonstrating that in the GT treated animals oxalurea reduced to normal levels with restoration of liver enzyme levels in the absence of any hepatotoxicity or immune reactions.

 

Clinical need:  Currently, most of the therapeutic options are diet-mediated to reduce the amount of glyoxylate intake and maximize the intake of vitamin B6.  The most effective treatment for PH1 is pre-emptive liver transplantation, alone or liver combined with kidney transplantation in ESRF.  There is therefore a clear need for alternative or new treatments options.

 

Feasibility :

 

Gene : the AGXT gene maps onto chromosome 2q36-q37, has a 10 kb coding sequence and contains 11 exons generating a 392-residue protein.

 

Vector optimization: Eight constructs different containing codon optimized AGXT genes have already been generated in-house and are currently being characterized.

 

Animal models: Small animal models already exist and have been used for pre-clinical proof of concept studies.

 

Biomarkers: Measurements of oxalate are part of routine clinical practice for the disease setting and monitoring of kidney changes can also be done using standard techniques.

 

After a phase of further vector optimization it is our aim to develop this product for a first Phase I clinical study by mid 2016. Further development timelines have not been defined.

 

Development overview to IMPD:

 

 

The GLP safety and toxicology studies will provide the first milestone (Go/No Go) for the project.

 



 

Safety Assessment: At this stage is not possible to make any inferences in relation to potential safety concerns.

 

C) Exploratory Research Projects

 

The projects listed under this category in Table 1 above are not in active research yet, but are likely targets for our platform technology and are being assessed on feasibility before starting active bench work.

 



 

Central Nervous System Programs

 

A)            Development Programs

 

1.               AMT-110 for Sanfilippo B

 

1.1.1         Disease Background

 

Sanfilippo syndrome, or Mucopolysaccharidosis type III (MPSIII), is a rare lysosomal storage disorder (LSD) that occurs when enzymes needed to break down the heparan sulfate sugar chain are missing or are defective. Sanfilippo B is one of the four types of MPSIII that results in serious brain degeneration in children, and is generally lethal. The deficient enzyme responsible for the disease is alpha-N-acetylglucosaminidase (NaGlu). The clinical manifestations are mainly neurological, with early symptoms observed during the first five years of age, leading to a progressive deterioration of cognitive abilities. Affected children require specific care after age seven and progressively develop profound mental retardation with reduced somatic manifestations. Death frequently occurs at the median age of 15. No treatment is currently available.

 

Birth prevalences of 0.28—4.1 per 100, 000 have been reported (Valstar et al., 2008). More recently, He´ron et al. (2010) estimated the mean annual incidence for Sanfilippo B in France at 0.15 per 100,000 births.

 

1.1.2         Overview of AMT-110

 

The goal of our AMT-110 program is to provide a gene therapy for Sanfilippo B syndrome through the introduction of a functional NaGlu gene into the patients’ brain cells.

 

This project is being pursued together with the Pasteur Institute (Paris) whereby uniQure is responsible for developing the manufacturing process and producing clinical grade material and the Pasteur Institute for conducting the clinical trials.

 

1.1.3         Preclinical Development

 

·                   Product Profile

 

AMT-110 is designed to be delivered via intracranial administration.

 

AMT-110 or rAAV5-hNaGlu, is a recombinant adeno-associated vector of serotype 5, consisting of:

 

·                   Inverted terminal regions or ITRs of the adeno-associated serotype 2

 

·                   A human α-N-acetylglucosaminidase, or hNaGlu, gene the therapeutic gene

 

·                   The mouse phosphoglycerate kinase-1 promoter (muPGK)

 

·                   Pre-clinical Proof of Concept

 

Preclinical PoC studies were conducted in mouse and dog disease models at the Pasteur Institute. These studies showed that mice with MSPIIIB a single AAV5-NaGlu intracranial injection

 



 

resulted in reversion of storage lesions throughout the brain and prevented loss of Purkinje cells. Furthermore, it improved animal behavior and corrected pathological featured of the disease including, neuro-inflammation, axonal transport, synaptic vesicle content and the autophagy defect.

 

In MSPIIIB dogs treatment (four simultaneous injections) with AAV5-hNaGlu was well tolerated, vector particles were broadly distributed and the therapeutic enzyme was delivered to the entire brain leading to improvement in MPSIIIB-induced histological lesions and normalization of glycosaminoglycan and ganglioside levels. These studies also confirmed that the combination of gene therapy with efficient immunosuppression was required for treatment efficacy.

 

·                   Non-clinical safety & toxicology studies

 

The following table presents a summary of the AMT-10 non-clinical safety and toxicology studies that have been conducted to support the clinical development program.

 

Parameter to be
assessed

 

Study performed

 

Results

Pharmacokinetics

 

1) GLP biodistribution in Sprague-Dawley rats with or without immunosuppression (180 days)

 

2) GLP biodistribution in Dogs with immunosuppression (90 days)

 

In in Sprague-Dawley rats:

·                        Constant high number of genome copies present in the brain with no vector in the blood at 3 months.

 

·                        No difference between immunosuppressed and non-immuno-suppressed animals.

In dogs

·                        Organs negative for vector DNA (< LOQ) were heart, lung, kidney and testis whereas positive organs (> LOQ) included spleen, liver and cervical spinal cord.

 

 

 

 

 

Toxicity

 

GLP toxicity study in Sprague-Dawley rats (180 days)

 

(French Authorities endorsed the use of a single species for safety and toxicology studies)

 

·                       No specific toxicity

 

·                        Only injection traces in the brain at some injection sites

 

·                        No effect on body weight, growth, food consumption, behaviour

 

·                        No decedents, no inflammation

 

·                        No histopathological findings

 

·                        NOAEL: 3.4 x 10 11 /animal

 

 

 

 

 

Persistence in semen and risk of

 

 

 

Based on pilot dog preliminary data where no vector DNA was found in the

 



 

germline transmission

 

 

 

gonads, French Authorities endorsed the proposal that no germline transmission studies were necessary.

 

1.1.4                               Summary of AMT-110 Preclinical Development Program

 

·                   In animal models of Sanfilippo B, treatment with AAV5-hNaGlu ameliorated pathophysiological signs and symptoms of the disease.

 

·                   AMT-110 administered into the striatum of non-immunosuppressed rats and immunosuppressed rats and dogs produced long lasting presence of vector DNA in the brain and caused no mortality and no signs of toxicity.

 

1.1.5

 

1.1.6         Clinical Development Program

 

1.1.7         The key regulatory and clinical development milestones for AMT-110 include the following,

 

·               1 st  Scientific Advice with French Regulatory Authorities

March 2012

 

 

·               2 nd  Scientific Advice with French Regulatory Authorities

March 2012

 

 

·               IMPD Submission

April 2013

 

 

·               IMPD Approval

3Q13

 

 

·               Phase I start

October 2013

 

Expected Milestones

 

·               Phase II/III start

2016

 

 

·               Registration

2018

 

The Phase I/II study is a single center, open label, prospective, interventional, single dose of AAV5-hFIXco (AMT-060) trial in children with Sanfilippo type B syndrome. Administration will be performed into the brain parenchyma and cerebellum at 8 image-guided tracks, with 2 deposits per tracks, in a single neurosurgical session. Each patient will receive 960 μL of vector suspension as 16 simultaneous vector deposit each containing 2.4 10 11  gc (4 10 12  gc in total). Patients will receive immunosuppression starting 10 days prior to treatment.

 



 

The primary objective of the study is to evaluate the clinical, radiological and biological safety of the treatment. The secondary objective is to collect samples and data to define exploratory tests that could become evaluation criteria for further clinical efficacy studies (Brain MRI; neurological tests and biological markers).

 

The study will be conducted at the Bicêtre Hospital which is part of the University Hospitals of South Paris and is expected to enroll a total of 4 children during an 8 to 12 months inclusion period. The duration of follow-up for each patient is one year. The first patient was dosed in October 2013.

 

·                   Future Clinical Development

 

Licensee plans to complete the Phase I and start a Phase II/III trial in multiple sites worldwide.  Following initiation of this trial one of the options on how to proceed would be applying for approval for compassionate use to treat on a named patient basis. This can be well justified based on the size of the indication and lethality of the condition.

 

1.1.8       Summary of AMT-110 Clinical Development Program

 

·                   The IMPD was submitted in April 2013

 

·                   Phase I was started in October 2013

 

B) Active Research Projects

 

1.               Huntington’s Disease

 

Disease background : Huntington’s Disease (HD) is a neurodegenerative genetic disorder that affects motor control and leads to cognitive decline and dementia. It typically becomes noticeable in middle age, but can begin at any age from infancy to old age. HD has a prevalence of around 1 affected individual in 100,000.

 

The mutated form of the protein huntingtin causes cellular dysfunction and death in a number of CNS sites but is most noticeable in the striatum and cortex. The mutation is caused by CAG repeats in the DNA of patients. The earliest features of HD are involuntary movements and irritability and a loss of executive function. This progresses over time and in the more advanced stages, the patient is demented and bed-bound. The disease is currently incurable with patients dying about 20-25 years after it begins.

 

Clinical need : The clinical need for these patients is high as there is no cure for the disease.

 

Feasibility

 

As the CAG repeats in the Huntingtin gene are the cause of the disease, downregulation of the expression of the CAG repeats is an option. Also rescuing the neurons from degeneration using GDNF is an option. Both options are currently under investigation. Replacing the gene is not an option as this is far too large to fit into an AAV vector.

 

Several transgenic mice models exist. Severity and time of onset are based on the number of CAG repeats in the model. Mostly used are the R6/1 and R6/2 transgenic models.

 

Preclinical work: Proof of concept using GDNF has been established in one laboratory. Licensee is currently trying to establish this with our own vector in the laboratory of Roger Barker.

 

Proof of concept with siRNA has been established in mice models and Licensee is in the process of implementing this into our studies.

 



 

Development overview to IMPD:

 

 

The proof of concept studies ( in vivo/ in vitro work) will provide the first milestone (Go/No Go) for the project.

 

With regards to the siRNA approach to HD, vector generation & optimization will require an additional 9 months prior to any other activity.  Then a similar development path to what is shown above will need to be followed.

 

It is Licensee’s aim upon a successful PoC to develop this product further to a Phase I clinical investigation which should start mid 2016.

 

Collaborators: Licensee is working together with Roger Barker (Cambridge University) on the use of GDNF to rescue neurons in Huntington models, based on a EUREKA grant. Licensee is also working together with Nicole Deglon (Lausanne University), Anna Skorupska (Lublin University) and Sebastian Kuegler (Gottingen University) in a Eurostars grant setting. Competition comes from siRNA companies.

 

Safety concerns: Potential safety concerns could be the complete downregulation of the Huntingtin gene, even though not fully supported by the Eurostars team. The use of GDNF could lead to side effects, such as weight loss.

 

IP: For GDNF, Licensee has a license from Amgen. For the siRNA work Licensee has a non-exclusive license from Benitec.

 

2.               Multiple System Atrophy

 

Disease Background: Multiple System Atrophy (MSA) is a sporadic neurodegenerative disease that is characterized by the presence of glial inclusion bodies, which stain positive for a synuclein. The clinical picture is that of parkinsonism, autonomic failure, cerebellar ataxia and pyramidal signs in differing combinations. Approximately 80% of patients present with predominantly parkinsonian features (MSA-P) manifesting in rapidly deteriorating akinesia, rigidity, postural instability and high pitched dysarthria.  Most such patients do not exhibit the classic resting tremor of Parkinson’s disease and virtually all develop frank dysautonomia in the course of the illness.  The cause of the disease is not known.

 

Clinical need: Although a minority of patients may achieve modest benefit from dopaminergic therapy, there is no satisfactory treatment for the parkinsonian disabilities of MSA-P. Additionally, deep brain stimulation of the subthalamic nucleus has been of little or no value. Within 5 years of disease onset patients die so the clinical need is high for these patients.

 

Feasibility:

 

MSA is not a single monogenic disease, but may be treated with a single neuroprotective protein. In this case, this could be GDNF. Some transgenic animal models exist, all overexpressing the alpha-synuclein protein. The rationale to use GDNF (besides its general neuroprotective effect on neurons) is that both in patients and the transgenic mouse model, GDNF expression is downregulated. Introduction of an elevated level of GDNF may

 



 

serve as the treatment. Read out parameters for the disease progression are all related to those of Parkinson’s Disease. PoC has not yet been established, but is under investigation in the mouse model.

 

Development overview to IMPD: a

 

 

The proof of concept studies ( in vivo/ in vitro work) will provide the first milestone (Go/No Go) for the project.  It is our aim upon a successful PoC to develop this product further to a Phase I clinical investigation which should start mid 2016.

 

Collaborators: Licensee is working together with Erwan Bezard (University of Bordeaux) and Olivier Rascol (University of Toulouse) who are together running the French reference center for MSA.

 

Safety Assessment: The use of GDNF could lead to side effects, such as weight loss. The exact mechanism through which the treatments would have its effect is not clear yet.

 

3.               Hearing loss

 

Disease background : Hearing loss is a serious clinical problem. Underlying mechanisms for the loss of neurons in the cochlea can vary from ischemia, mechanical stress to toxic insults. The actual numbers of patients is not easy to define, but it could be rather large. When age-related hearing loss is also taken into account, this is no longer an orphan indication.

 

Clinical need : Patients with hearing loss could be helped with cochlear implants. However, progressive neurodegeneration is not stopped by that. There is high clinical need as there is no cure for the disease.

 

Feasibility:

 

Neuron function and survival is dependent on a delicate balance of neurotrophins. Following trauma or toxic insult to neurons, they may slowly die. To reverse this state of degeneration, it could be beneficial to supply the neurons with a neurotrophin such as GDNF. This neurotrophin has been shown to be able to rescue neurons from degeneration in several models, including those of the substantia nigra and for instance motorneurons in the spinal cord after trauma.

 

Animal models are available and include for instance use of Kanamycin in cats, mice or guinea pigs. Also chemotherapeutic agents from the class of statins are used.

 

Preclinical work: Proof of concept using recombinant brain-derived neurotrophic factor (BDNF) and/or GDNF has been established. Licensee is currently trying to establish this with our own vector in the laboratory of Patricia Leake.

 

Cochlea of mice can be transduced to express a recombinant transgene.

 



 

Development overview to IMPD:

 

 

The proof of concept studies ( in vivo/ in vitro work) will provide the first milestone (Go/No Go) for the project.

 

This new project has just been initiated upon a successful PoC it is our aim to develop this product further to a Phase I clinical trial, which should start by the end 2016.

 

Collaborators: Licensee is working together with Patricia Leake (University College of San Francisco) on the use of GDNF to rescue neurons in mouse and cat models. She is the investigator who developed the cochlear implant. This could also be included in the experimental plan.

 

Safety concerns: The use of GDNF could lead to side effects. Weight loss is not expected, but as the GDNF also has a neurotrophic effect, nerve fibers could sprout in an aberrant way possibly leading to incorrect connections.

 

IP: For GDNF, Licensee has a license from Amgen; the program as a whole is under investigation.

 

C) Exploratory Research Projects

 

The projects listed under this category in Table 1 above are not in active research yet, but are likely targets for our platform technology and are being assessed on feasibility before starting active bench work.

 


Exhibit 10.3

 

EXCLUSIVE LICENSE AGREEMENT

 

THIS Exclusive License Agreement (“Agreement”), effective as of 07 July 2008 (“Effective Date”), is entered into by and between St. Jude Children’s Research Hospital, Inc., a Tennessee not-for-profit corporation located at 262 Danny Thomas Place, Memphis, Tennessee 38105 (“ST. JUDE”) and Amsterdam Molecular Therapeutics B.V., a closed limited liability company organized and existing under the laws of the Netherlands, with registered offices at Meibergdreef 61, 1105 BA Amsterdam, the Netherlands, (each a “Party” and together the “Parties”).

 

RECITALS

 

WHEREAS, ST. JUDE is the owner by assignment from Dr. John Gray of his entire right, title and interest in the Patent Rights and in the inventions described and claimed therein; and

 

WHEREAS, the Patent Rights relate to mechanisms for improving the expression of Factor IX in gene therapy vectors, including, the use of a specific Factor IX polynucleotide coding sequence designed for optimal expression, and, the use of transcriptional regulatory regions minimized in size so that they can be used to express Factor IX, as well as any other gene of interest, in a size-constrained environment such as in a self complementary gene therapy vector system; and

 

WHEREAS, under a separate agreement being entered into between the Parties on the same date as this Exclusive License Agreement (“Sponsored Research Agreement”), AMT engages ST. JUDE to develop a gene therapy vector incorporating certain features of the invention described in the Patent Rights but combined with certain technologies controlled by AMT (the “Vector”); and

 

WHEREAS, in order to carry out the work anticipated under the Sponsored Research Agreement, ST. JUDE will require access to technologies owned by AMT and AMT is willing to grant this access under a separate agreement being entered into between the Parties on the same date as this Exclusive License Agreement (“AMT Technology License Agreement”); and

 

SIGNATURE VERSION

 



 

WHEREAS, the Vector will be further developed and commercialized by AMT and so AMT require a license under the Patent Rights; and

 

WHEREAS, AMT wishes to enter into an agreement with ST. JUDE to obtain a license under the Patent Rights and ST. JUDE is willing to grant such a license to AMT under the terms and conditions set forth in this Agreement.

 

NOW, THEREFORE, in consideration of the mutual promises contained herein and other good and valuable considerations, the Parties do hereby agree as follows:

 

1.                                       DEFINITIONS

 

1.1                                “Affiliate” shall mean any company, partnership or other business entity which Controls, is Controlled by or is under common Control with either Party.  For the purposes of this definition “Control” means any of the following: (i) the possession, directly or indirectly, of the power to direct the management or policies of an entity, whether through ownership of voting securities, by contract or otherwise; (ii) ownership of fifty percent (50%) or more of the voting securities entitled to vote for the election of directors in the case of a corporation, or of fifty percent (50%) or more of the equity interest in the case of any other type of legal entity; (iii) status as a general partner in any partnership, or any other arrangement whereby a Party controls or has the right to control the board of directors or equivalent governing body of a corporation or other entity.

 

1.2                                “AMT” shall mean Amsterdam Molecular Therapeutics B.V. and any Affiliates of Amsterdam Molecular Therapeutics B.V.

 

1.3                                “Confidential Information” shall mean any confidential or proprietary non-public information furnished by one Party (the “Disclosing Party”) to the other Party (the “Receiving Party”) in connection with this Agreement.

 



 

1.4                                “Commercialise”, “Commercialisation” or “Commercialising” shall mean all activities relating to the import, advertising, promotion and other marketing, pricing and reimbursement, detailing, distribution, storage, handling, offering for sale and selling, customer service and support or post regulatory approval regulatory activities in relation to Licensed Product.

 

1.5                                “Commercially Reasonable Efforts” shall mean efforts and resources commonly used by biotechnology companies of a similar size to AMT based on funds raised to Develop and Commercialise a product owned by such a company or to which it has rights, which product is at a similar stage in its development or product life and is of similar market potential to the Licensed Product in question and taking into account the patent and other proprietary position of the product.

 

1.6                                “Development (and the corresponding verb “to Develop”)” shall mean all pre-regulatory approval development and regulatory activities regarding a product in a country conducted with the aim of obtaining such regulatory approval including:

 

(a)                                  studies on the toxicological, pharmacological, metabolical or clinical aspects of a product conducted internally or by individual investigators or consultants; and

 

(b)                                  process development, process improvement, scale-up and recovery, the manufacture of clinical supplies of product, including failed batches, manufacture of qualification lots; and

 

(c)                                   preparing, submitting, reviewing or developing data or information for the purpose of submission to a regulatory authority to obtain, maintain and/or expand manufacturing and/or regulatory approval of a product including data management, statistical designs and studies, document preparation, and other administration; and

 



 

(d)                                  all post regulatory approval regulatory activities and adverse event reporting in relation to products.

 

1.7                                “Field” shall mean gene therapy for the therapy or prophylaxis of Haemophilia B.

 

1.8                                “Investigational New Drug Application (“INDA”) shall mean an application submitted to the FDA or a foreign equivalent requesting permission to conduct human clinical studies with an investigational new drug or to conduct human clinical studies with an existing drug for a new use.

 

1.9                                “Licensed Product” shall mean all products containing Factor IX that are covered by Valid Claims of the Patent Rights to any extent.

 

1.10                         “Net Sales” shall mean the sum of all amounts actually invoiced by AMT (or Sublicensees as appropriate) from persons or entities for sales of Licensed Products, less the following deductions and offsets, to the extent such sums are actually incurred, paid or credited by AMT (or Sublicensees as appropriate) and not otherwise reimbursed:

 

(a)                                  normal and customary trade, cash and quantity discounts actually given, credits, price adjustments or allowances for damaged products, returns or rejections of products;

 

(b)                                  chargeback payments and rebates (or the equivalent thereof) for product granted on a customary trade basis to group purchasing organisations, managed health care organisations or to federal, state/provincial, local and other governments, including their agencies, or to trade customers;

 

(c)                                   reasonable and customary freight, shipping insurance and other transportation expenses directly related to the sale of product (if actually borne by AMT or Sublicensees without reimbursement from any third party);

 



 

(d)                                  required distribution commissions/fees payable to any third party providing distribution services to AMT or Sublicensees;

 

(e)                                   sales, value-added, excise taxes, tariffs and duties, and other taxes and government charges directly related to the sale, to the extent that such items are included in the gross invoice price of product and are actually borne by AMT or its Sublicensees without reimbursement from any third party (but not including taxes assessed against the income derived from such sale); and

 

(f)                                    actual uncollectible amounts for product where collectibility is determined in accordance with IFRS consistently applied to all AMT products.

 

Sales of products intended for resale to third parties, and made internally amongst AMT and Sublicensees shall not be deemed sales for purposes of calculating “Net Sales” (however, sales to a third party other than a Sublicensee or Affiliate shall be included in the calculation of “Net Sales”).

 

1.11                         “Patent Rights” shall mean (i) U.S. Provisional Patent Application No. 60/612,135 filed September 22, 2004 entitled “Improved Expression of Factor IX in Gene Therapy Vectors” (ST. JUDE reference no. SJ-04-0024); (ii) all patent applications filed claiming priority from the above including all provisional patent applications and all national, regional and international patents and patent applications; (iii) all patent applications filed claiming priority from any of the above including any divisional, continuation, or continuation-in-part; (iv) any patent issued on any of the foregoing; (v) any reissue or extension of such patent; and (vi) any foreign counterparts to such patents and patent applications and applications and/or patents or the equivalent thereof claiming priority to or from any of the above.

 

1.12                         “Phase I Clinical Trial” shall mean a human clinical trial, the principal purpose of which is a preliminary determination of safety in healthy individuals or patients as required in 21

 



 

C.F.R. §312, or a similar clinical study prescribed by the regulatory authorities in a country other than the United States.

 

1.13                         “Phase II Clinical Trial” shall mean (i) a human clinical trial, for which a primary endpoint is a preliminary determination of efficacy or dose ranges in patients with the disease target being studied as required in 21 C.F.R. §312.21(b), as maybe amended from time to time, or a similar clinical study prescribed by the regulatory authorities in a country other than the United States, or (ii) a combined Phase II and Phase III Clinical Trial which enrolls at least forty (40) patients, or any Phase III Clinical Trial performed in lieu of a Phase II study.

 

1.14                         “Phase III Clinical Trial” shall mean a human clinical trial, the principal purpose of which is to establish safety and efficacy in patients with the disease target being studied as required in 21 C.F.R. §312, or similar clinical study prescribed by the regulatory authorities in a country other than the United States.  A Phase III Clinical Trial shall also include any other human clinical trial intended as a pivotal study, whether or not such study is a traditional Phase III study.

 

1.15                         “Royalty Reporting Period” shall mean the partial calendar half year commencing on the date on which a Licensed Product is first sold and every complete or partial calendar half year thereafter during which royalty payment obligations under this Agreement remain in effect.

 

1.16                         “Sublicensee” shall mean any sublicensee of the rights granted to AMT under this Agreement, as further described in Section 2.1(i).

 

1.17                         “ST. JUDE Improvements” shall mean any improvement or enhancement (whether patentable or not) to the inventions of a Valid Claim of the Patent Rights in the Field that is discovered, developed or otherwise acquired by ST. JUDE after the Effective Date

 



 

pursuant to Section 2.1(v) (but, for clarity, outside the scope of the Sponsored Research Agreement) and in relation to which a patent application is filed by ST. JUDE.

 

1.18                         “Valid Claim” shall mean a claim of an issued and unexpired patent or pending published patent application included within Patent Rights, which has not been held permanently revoked, unenforceable or invalid by a decision of a court or other governmental agency of competent jurisdiction, unappealable or un-appealed within the time allowed for appeal, and which has not been admitted to be invalid or unenforceable through reissue or disclaimer or otherwise.

 

2.                                       GRANT

 

2.1                                License Grant

 

(i)                                      ST. JUDE hereby grants to AMT and AMT accepts, subject to the terms and conditions herein, an exclusive worldwide license under the Patent Rights to research, have researched, Develop, have Developed, make, have made, import, distribute, use and Commercialise Licensed Products in the Field.  Such license shall include the right to grant sublicenses provided that AMT shall remain responsible for compliance by Sublicensees with the terms and conditions of this Agreement.  Within thirty (30) days of the grant of each sublicense under this Agreement, AMT shall inform ST. JUDE in writing of the identity of the Sublicensee and provide a copy of the sublicense agreement but showing only those terms directly pertaining to the sublicense itself, with all other terms including financial terms redacted.

 

(ii)                                   The license granted herein is subject to the rights, conditions and limitations imposed by U.S. law on inventions and discoveries conceived or first actually reduced to practice during the course of research funded by a U.S. federal agency that are relevant to the Patent Rights.  The words “exclusive license” as used herein

 



 

shall mean exclusive except for the royalty-free non-exclusive license granted to the U.S. government by ST. JUDE pursuant to 35 USC Section 202 (c) (4) for any Patent Rights claiming any invention subject to such license as defined under 35 USC Section 201 and any other federal laws and applicable regulations.

 

(iii)                                To the extent that Licensed Products embody Patent Rights conceived or first actually reduced to practice during the course of research funded by a U.S. federal agency, AMT agrees that such Licensed Products shall be manufactured substantially in the United States in accordance with 35 U.S.C. Section 204.

 

(iv)                               Title to the Patent Rights shall remain with ST. JUDE and ST. JUDE retains the right to license the Patent Rights to other entities outside the Field and to use the Patent Rights for internal and collaborative research outside the Field.

 

(v)                                  The license granted under Section 2.1(i) of this Agreement is subject to the non-transferable right of ST. JUDE under the Patent Rights solely to perform internal and collaborative research and education in the Field with academic collaborators.  To the extent that such research involves pre-clinical or clinical research the data and other results of such research, including any IND package and a copy of any interim or final clinical research report shall be made available by ST. JUDE to AMT and AMT shall be permitted to utilise the same only for lawful purposes in its dealings with the FDA.  ST. JUDE will at its own cost procure that this is possible under the terms of any agreement between it and such academic collaborations.

 

2.2                                ST. JUDE Improvements.  If any ST. JUDE Improvements are made by ST. JUDE during the term of this Agreement, AMT shall have the first right of refusal to such ST. JUDE Improvements.  ST. JUDE will disclose any such ST. JUDE Improvement to AMT by notice in writing.  AMT shall treat any such disclosure as Confidential Information and

 



 

shall only use such disclosure to consider its licensing interest.   AMT shall have sixty (60) days from the date of such disclosure in which to indicate to ST. JUDE by notice in writing whether it wishes to negotiate a license with grant terms the same as those set out in Section 2.1.  If AMT does so indicate that it wishes to take a license within the notice period, the Parties shall negotiate in good faith the financial and other terms of such license during the subsequent period of ninety (90) days.  If the Parties cannot reach agreement during the negotiation period, ST. JUDE will be free to deal with third parties in respect of the said ST. JUDE Improvements in the Field.  During the period from first notification by ST. JUDE of the ST. JUDE Improvement to AMT until the expiration of the aforesaid ninety (90) day period, the ST. JUDE shall not enter into arrangements or agreements with any third party concerning the ST. JUDE Improvements in the Field.

 

3.                                       DILIGENCE OBLIGATIONS AND ANNUAL PROGRESS REPORT

 

3.1                                Use of Commercially Reasonable Efforts.  With effect from completion or termination of the Research Program under the Sponsored Research Agreement (“Commencement Date”) AMT shall use Commercially Reasonable Efforts to diligently Develop and Commercialize Licensed Products whether by itself or through its Sublicensee(s).

 

3.2                                Annual Progress Reports.  Within thirty (30) days after each anniversary of the Commencement Date, AMT shall furnish ST. JUDE with a written report summarizing efforts and achievements toward Developing and Commercializing Licensed Products, including the status of any regulatory submissions, clinical trials and sublicensing activities.  This report shall also include a statement regarding insurance coverage in accordance with Section 8.1 (iii) below.

 

4.                                       PAYMENTS

 

4.1                                License Fee.  In partial consideration of the rights granted to AMT under this Agreement and to reimburse ST. JUDE for patent expenses already incurred in pursuing the Patent

 



 

Rights, AMT shall pay Forty Five Thousand ($45,000) U.S. dollars to ST. JUDE within fifteen (15) days of the full execution of this Agreement.  This license fee payment is nonrefundable and not creditable against any other payments due to ST. JUDE under this Agreement.

 

4.2                                Annual Maintenance Fee.  AMT shall pay ST. JUDE an annual fee of Ten Thousand ($10,000) U.S. dollars within thirty (30) days of January 1.  This fee shall be creditable to royalties and milestones which are due in the same calendar year.

 

4.3                                Milestone Payments.  AMT shall pay ST. JUDE the following milestone payments on the first occurrence of the following milestone events:

 

(i)                                      Five Hundred Thousand ($500,000) U.S. dollars upon dosing of the first patient with a Licensed Product in a Phase I Clinical Trial;

 

(ii)                                   One Million ($1,000,000) U.S. dollars upon dosing of the first patient in a Phase III Clinical Trial or equivalent clinical trial of a Licensed Product; and

 

(iii)                                Five Million ($5,000,000) U.S. dollars upon the grant of regulatory marketing approval for a Licensed Product in the U.S. or Europe.

 

Each of the milestone payments the subject of this Section shall only be payable by AMT upon the first occurrence of the applicable event whenever it occurs.

 

4.4                                Royalties.  AMT shall pay ST. JUDE two percent (2%) of Net Sales of Licensed Products sold by AMT itself or Sublicensees on a country by country basis until expiry of the Valid Claims of the Patent Rights in the country of sale that cover the product and render it a Licensed Product.

 



 

4.5                                Sublicense Consideration.  In addition to the royalty obligation as set forth under Section 4.4, AMT shall pay to ST. JUDE the following percentages of consideration received for sublicenses under this Agreement:

 

(i)                                      Fifteen percent (15%) for a sublicense granted after AMT has completed preclinical research but before completion of a Phase I Clinical Trial for a Licensed Product;

 

(ii)                                  Ten percent (10%) for a sublicense granted after completion of a Phase I Clinical Trial for a Licensed Product but before completion of a Phase II Clinical Trial; and

 

(iii)                               Five percent (5%) for a sublicense granted after completion of a Phase II Clinical Trial for a Licensed Product.

 

This payment shall be due, without the need for invoice from ST. JUDE, within forty-five (45) days of the receipt the payment made to AMT by a Sublicensee under a sublicense agreement.  Such sublicense consideration shall include consideration of any kind received by AMT from a Sublicensee for the grant of a sublicense under this Agreement, such as upfront fees or milestone fees and including any premium paid by the Sublicensee over the Fair Market Value (as such term is defined in subsection (iii) below) for stock of AMT in consideration for such sublicense. However, not included in such sublicense consideration are:

 

(i)                                      Support for research, Development and/or manufacturing activities corresponding directly to the Development of Licensed Products, which do not exceed the fully-burdened cost for undertaking such research, Development, and/or manufacturing performed by or for AMT (including third parties on AMT’s behalf), each pursuant to a specific agreement including a performance plan and commensurate budget;

 



 

(ii)                                  Proceeds derived from debt financing, to the extent that such financing is at market rates, and any loans to AMT by the Sublicensee:

 

(iii)                              Consideration received for the purchase of an equity interest in AMT to the extent that the price per share for such equity does not exceed by more than twenty-five percent (25%) the Fair Market Value of AMT’s stock.  The term Fair Market Value shall mean the average price that the stock in question is publicly trading at for twenty (20) days prior to the announcement of its purchase by the Sublicensee or if the stock is not publicly traded, the value of such stock as determined by the most recent private financing through a financial investor (an entity whose sole interest in AMT is financial);

 

(iv)                              Reimbursement of AMT’s patent costs related to Patent Rights; and

 

(v)                                 Any and all amounts paid to AMT by a Sublicensee as royalties on sales of Licensed Product sold by the Sublicensee under a sublicense.

 

4.6                                Late Payment.  For any late payment AMT shall pay an interest penalty based on the amount owed at a daily accrual rate equal to the lesser of ten percent (10%) per annum or the highest rate permissible by law.

 

5.                                       ROYALTY REPORTS; PAYMENTS; RECORDS

 

5.1                                First Sale.  AMT shall report to ST. JUDE the date of first commercial sale of a Licensed Product within thirty (30) days of its occurrence.

 

5.2                                Reports and Payments.  Within sixty (60) days after the conclusion of each Royalty Reporting Period, AMT shall deliver to ST. JUDE a report of Net Sales for each Licensed Product during the prior Royalty Reporting Period on a country-by-country basis. Such report shall include the amount of gross sales and the amount of all deductions and

 



 

reductions taken in each category identified in the definition of Net Sales in sufficient detail to allow ST. JUDE to verify the Net Sales calculation, the amount of Net Sales, and the total royalty payable on Net Sales in U.S. dollars, together with the exchange rates used for conversion.  All such reports shall be considered Confidential Information of which AMT is the Disclosing Party and the provisions of Section 7 of this Agreement shall apply to such reports.  If no royalties are due to ST. JUDE for any Royalty Reporting Period, the report shall so state. Concurrent with the report, AMT shall remit to ST. JUDE any payment due for the applicable Royalty Reporting Period. Unless other arrangements are made, payment shall be remitted to the following address:

 

St. Jude Children’s Research Hospital

P.O. Box 1000, Department # 516

Memphis, TN 38148-0516

 

5.3                                Records.

 

(i)                                      AMT shall maintain, and shall require its Sublicensees to maintain complete and accurate records of all Net Sales under this Agreement which records shall contain sufficient information to permit auditors to confirm the accuracy of any reports delivered to ST. JUDE under Section 5.2.  The relevant party shall retain such records relating to a given Royalty Reporting Period for at least three (3) years after the conclusion of that Royalty Reporting Period.

 

(ii)                                  Upon twenty (20) working days notice by ST. JUDE, ST. JUDE shall have the right, at its expense, to cause accountants from a nationally-recognized accounting firm to inspect the records of AMT (but not Sublicensees) for the period covering Royalty Reporting Periods ending no more than three (3) years prior to the date of the inspection (including records of royalty accounting received from their Sublicensees) during normal business hours for the sole purpose of verifying any

 



 

reports and payments delivered under this Section 5.2. ST. JUDE may exercise its rights under this Section 5.3 only once every calendar year.

 

(iii)                               The Parties shall reconcile any underpayment or overpayment within thirty (30) days after the auditor delivers the results of the audit. In the event that any audit performed under this Section reveals an underpayment in excess of five percent (5%) in any Royalty Reporting Period, AMT shall bear the full cost of such audit.

 

(iv)                              Prior to any such audit taking place, such firm of accountants shall undertake to AMT that they shall keep all information and data contained in the records of AMT strictly confidential and shall not disclose such information or copies of such records to any third person including ST. JUDE, but shall only use the same for the purpose of the reviews and/or calculations which they need to perform in order to verify the reports delivered under Section 5.2 of this Agreement.

 

(v)                                 Upon timely request by ST. JUDE, AMT shall, at the expense of ST. JUDE, have any Sublicensee accounting to AMT for royalties audited under the audit provisions agreed between AMT and the Sublicensee, and AMT shall report to ST. JUDE the outcome. If there is a discrepancy identified upon such an audit the provisions of Section 5.3(iii) shall apply in like manner.

 

6.                                       PATENTS AND INFRINGEMENT

 

6.1                                Responsibility for Patent Rights.  Title to all Patent Rights shall remain with ST. JUDE and ST. JUDE shall retain primary responsibility for the drafting, filing, prosecution, and maintenance of all Patent Rights.  ST. JUDE shall appoint and retain external patent counsel approved by AMT.  ST. JUDE shall keep AMT informed of all developments including promptly furnishing AMT with all patent office communications.  ST. JUDE shall, to the satisfaction of AMT, implement all reasonable requests of AMT with respect to the drafting, filing, prosecution, and maintenance of all Patent Rights.

 



 

6.2                                Reimbursement of Patent Expenses.  AMT shall reimburse ST. JUDE for all out of pocket patent-related expenses incurred by ST. JUDE pursuant to Section 6.1 related to Patent Rights during the term of this Agreement within thirty (30) days after ST. JUDE invoices AMT subject to the provisions of this Section.  Such invoice shall contain a breakdown of the expenses and be accompanied by supporting evidence of such expenses as appropriate including copies of invoices from external patent counsel.  ST JUDE shall instruct its patent counsel to communicate with AMT directly on all matters pertaining to the activities of patent counsel, including the giving of forward cost estimates, but copying ST JUDE on all e-mails and other correspondence.

 

6.2.1                      If ST. JUDE grants an exclusive license under the Patent Rights outside of the Field, ST. JUDE shall promptly notify AMT. In such an event, from the effective date of that license, AMT shall be responsible for seventy five percent (75%) of the patent-related expenses only.

 

6.3                                Abandonment.  AMT may elect, upon sixty (60) days written notice to ST. JUDE, to cease payment of the expenses associated with obtaining or maintaining patent protection for one or more patents or applications within the Patent Rights in one or more countries.  In such event, AMT shall forfeit all rights under this Agreement with respect to such patent within the Patent Rights in such country(ies). ST. JUDE shall have the right, at its sole expense, to prepare, file, prosecute, and maintain any patents or patent applications under Patent Rights abandoned by AMT.

 

6.4                                Infringement.

 

(i)                                      Notification of Infringement.  Each Party agrees to provide written notice to the other Party promptly (i) after becoming aware of or having a reasonable suspicion of any infringement of the Patent Rights in the Field or (ii) upon becoming aware of any action alleging invalidity or non-infringement of the Patent Rights in the Field.

 



 

(ii)                                  AMT Right to Enforce Patent Rights in the Field.  AMT shall have the right, under its own control and at its own expense, to prosecute any third party infringement of the Patent Rights in the Field including negotiating sublicense agreements with such third parties at its discretion.  At least thirty (30) days prior to a potential claim, AMT shall notify ST. JUDE in writing of the nature of the anticipated action and the party(ies) against whom the anticipated action may be taken.  It is understood that any sublicense rights granted hereunder shall not forgive any royalty payments that would otherwise be due to ST. JUDE based on sales of Licensed Products by the Sublicensee without consultation with ST. JUDE.  If AMT succeeds in any such infringement proceedings whether at trial or by way of settlement, any recovery by AMT in such proceedings brought by AMT shall first be used to reimburse AMT for all reasonable out-of-pocket costs and legal fees incurred to conduct such proceedings.  Out of any remaining damages actually received by AMT relating to infringement of the Patent Rights, AMT shall pay to ST. JUDE an amount equivalent to the payment which would have been due to ST. JUDE on the balance as if they were Net Sales, along with an accounting of the recovery and the reasonable out-of-pocket costs and legal fees.

 

In the event that AMT fails to initiate an infringement action within 18 months after it first becomes aware of such infringement or notifies ST. JUDE that it does not intend to initiate such action, ST. JUDE shall have the right to prosecute such infringement, under its sole control and its sole expense, and any recovery obtained shall be retained by ST. JUDE. AMT shall provide all necessary assistance to ST. JUDE in relation to such proceedings and ST. JUDE shall on demand by AMT indemnify AMT against the costs of such activity, unless AMT elects to be separately represented (which shall be at AMT’s discretion), in which case such separate representation shall be at AMT’s cost and expense.

 



 

(iii)                               ST. JUDE as Indispensable Party.  If, in the reasonable opinion of AMT’s counsel, ST. JUDE should be a named party to any such suit, AMT may name ST. JUDE as a party, provided that AMT shall hold ST. JUDE harmless from, and if necessary indemnify ST. JUDE against, any costs (including attorney fees), expenses or liability that ST. JUDE may incur in connection with such action unless ST. JUDE elects to be separately represented in which case such separate representation shall be at ST. JUDE’s own cost and expense.

 

(iv)                              Cooperation.  Each Party agrees to cooperate fully in any action under this Section 6.4, which is controlled by the other Party, provided that the controlling Party reimburses the cooperating Party promptly for any costs and expenses incurred by the cooperating Party in connection with providing such assistance.

 

6.5                                Third Party Patent Rights.  Each Party shall promptly notify the other Party of any third party patent rights relevant to the Development or Commercialisation of the Licensed Product of which it becomes aware including by making appropriate searches for these as and when it considers appropriate.  To the extent possible, whilst preserving attorney-client privilege, the Parties’ patent counsel shall share copies of all external and internal opinions on the likelihood of grant and/or validity of relevant third party patent rights

 

7.                                       CONFIDENTIAL INFORMATION; PUBLICITY

 

7.1                                Confidential Information.

 

(i)                                     Obligations.  Except to the extent authorized in Section 7.1(i) of this Agreement, and for so long as the exceptions set out below in Section 7.1(ii) do not apply, the Receiving Party shall, in relation to any Confidential Information (i) maintain such Confidential Information in confidence using the same duty of care it would use to protect its own information of a like kind (and in any event no less than reasonable care), except that the Receiving Party may disclose or permit the disclosure of any

 



 

Confidential Information to its Sublicensees (to the extent necessary to effect the relevant Sublicense) and its trustees, directors, officers, employees, consultants, and advisors who are obligated to maintain the confidential nature of such Confidential Information and who need to know such Confidential Information for the purpose of this Agreement and for other purposes that may be required or necessary pursuant to this Agreement such as communication with collaborators or regulatory agencies; (ii) use such Confidential Information solely for the purposes of this Agreement; and (iii) allow its Sublicensees, trustees or directors, officers, employees, consultants, and advisors to reproduce the Confidential Information only to the extent necessary for the purposes of this Agreement, with all such reproductions being considered Confidential Information.

 

(ii)                                  Exceptions.  The obligations of the Receiving Party under Section 7.1(i) above shall not apply to the extent that the Receiving Party can demonstrate by written evidence that certain Confidential Information (a) was in the public domain prior to the time of its disclosure under this Agreement; (b) entered the public domain after the time of its disclosure under the Agreement through means other than an unauthorized disclosure resulting from an act or omission by the Receiving Party; (c) was independently developed or discovered by the Receiving Party without use of the Confidential Information; (d) is or was disclosed to the Receiving Party at any time, whether prior to or after the time of its disclosure under this Agreement, by a third party having no fiduciary relationship with the Disclosing Party and having no obligation to confidentiality with respect to such Confidential Information; or (e) was previously known to the Receiving Party from sources other than the Disclosing Party at the time of disclosure under this Agreement other than under an obligation of confidentiality.

 



 

(iii)                               Allowed Disclosure.  Notwithstanding the above obligations of confidentiality and non-use a Receiving Party may:

 

(a)                                  disclose Confidential Information to a competent authority as reasonably necessary to obtain regulatory approval in a particular jurisdiction to the extent consistent with the licenses granted under terms of this Agreement; and

 

(b)                                  disclose Confidential Information: (i) to the extent such disclosure is reasonably necessary to comply with the order of a court; or (ii) to the extent such disclosure is required to comply with a legal requirement, including to the extent such disclosure is required in publicly filed financial statements or other public statements under rules governing a stock exchange (e.g., EURONEXT, the rules of the United States Securities and Exchange Commission, NASDAQ, NYSE, or any other stock exchange on which securities issued by either Party may be listed); provided, to the extent possible bearing in mind such legal requirements and subject to the next sentence of this Section, such Party shall provide the other Party with a copy of the proposed text of such statements or disclosure five (5) Business Days in advance of the date on which the disclosure is to be made to enable the other Party to review and provide comments, unless a shorter review time is agreed.  If the compliance with a legal requirement requires filing of this Agreement, the filing Party shall to the extent possible seek confidential treatment of portions of this Agreement from the relevant competent authority and shall provide the other Party with a copy of the proposed filings at least ten (10) Business Days prior to filing for the other Party to review any such proposed filing.  Each Party agrees that it will obtain its own legal advice with regard to its compliance with legal

 



 

requirements and will not rely on any statements made by the other Party relating to such legal requirements; and

 

(c)                                   disclose Confidential Information by filing or prosecuting the Patent Rights, the filing or prosecution of which is contemplated by this Agreement, without violating the above confidentiality provisions; it being understood that publication of such filings occurs in some jurisdictions within eighteen (18) months of filing; and

 

(d)                                  in the case where AMT is the Receiving Party disclose Confidential Information to AMT’s contractors (including clinical researchers) distributors, Sublicensee’s, agents, consultants, as such Receiving Party reasonably determines is necessary to receive the benefit of the licenses and rights granted or available to it under this Agreement or to fulfil its obligations pursuant to this Agreement; provided, however, any such persons must be obligated to substantially the same extent as set forth in this Section to hold in confidence and not make use of such Confidential Information for any purpose other than that permitted by this Agreement; and

 

(e)                                   disclose Confidential Information: (i) to its actual or potential investment bankers; (ii) to existing and potential investors in connection with an offering or placement of securities for purposes of obtaining financing for its business and to actual and prospective lenders for the purpose of obtaining financing for its business; and (iii) to a bona fide potential acquiror or merger partner for the purposes of evaluating entering into a merger or acquisition, provided, however, any such persons must be obligated to substantially the same extent as set forth in this Section to hold

 



 

in confidence and not make use of such Confidential Information for any purpose other than those permitted by this Agreement; and

 

(f)                                    disclose Confidential Information to its legal advisers for the purpose of seeking advice.

 

7.2                                Use of Names.

 

(i)                                      AMT and its Sublicensees shall not use the name “St. Jude Children’s Research Hospital” or any variation of that name, or any trademarks or logos belong to ST. JUDE, or the names of any of ST. JUDE’s trustees, officers, faculty, students, employees, or agents, or any adaptation of such names, or any term of this Agreement in any promotional material or other public announcement or disclosure or in connection with the marketing or sale of any Licensed Product without the prior written approval of ST. JUDE; except: (a) in annual reports or as part of required regulatory or financial disclosures to the FDA, Securities Exchange Commission or other federal or foreign agencies; and (b) where otherwise required by law, provided that AMT shall notify ST. JUDE in advance of any disclosure to be made under these exceptions.

 

(ii)                                   ST. JUDE shall not use the name “ Amsterdam Molecular Therapeutics B.V.” or any variation of that name, or any term of this Agreement in any promotional material or other public announcement or disclosure without the prior written approval of AMT; except: (a) as part of required reports to state or federal government entities; and (b) where otherwise required by law, provided that ST. JUDE shall notify AMT in advance of any disclosure to be made under these exceptions.

 

7.3                                Publication.  If ST. JUDE wishes to publish or otherwise publically disclose results obtained from its internal research under the Patent Rights in the Field, ST. JUDE shall

 



 

submit to AMT a confidential copy of the intended publication or disclosure at least thirty (30) days prior to the proposed publication or other disclosure date.  If, AMT believes that the publication or intended disclosure contains patentable subject matter or contains Confidential Information of AMT, AMT shall notify ST. JUDE in writing.

 

7.4                                Marking.  AMT shall mark all Licensed Products intended for use under Patent Rights with appropriate information with respect to Patent Rights in accordance with the statutes of the United States relating to the marking of patented articles (see 35 U.S.C. §287(a)) and corresponding foreign rules and regulations.

 

8.                                       INDEMNIFICATION AND INSURANCE

 

8.1                                Indemnification.

 

(i)                                      Indemnity.

 

(a)                                  Except with respect to third party claims the subject of this Section, neither Party shall be liable to the other in contract, tort, negligence, breach of statutory duty or otherwise for any loss, damage, costs or expenses of any nature whatsoever incurred or suffered by the other or its Affiliates of a direct nature where the same is a loss of turnover, profits, business or goodwill; or an indirect or consequential or punitive nature, including any indirect or consequential economic loss or other indirect or consequential loss of turnover, profits, loss of enterprise value, business or goodwill or otherwise.

 

(b)                                  AMT shall indemnify, defend and hold ST. JUDE, the American Lebanese Syrian Associated Charities, Inc. (ALSAC; a non-profit, 501(c)(3) corporation which supports ST. JUDE), their present and former trustees, directors, governors, officers, agents, faculty, employees and students (“the

 



 

Indemnitees”) harmless as against any claims, demands, damages, judgments, fees (including reasonable attorneys fees), expenses, or other costs arising from or incidental to any product liability or other lawsuit, claim, demand or other action brought by a third party as a consequence of the use of clinical data provided by ST. JUDE, the practice of the Patent Rights or the sale of Licensed Products by AMT or Sublicensees, whether or not ST. JUDE, either jointly or severally, is named as a party defendant in any such lawsuit and whether or not ST. JUDE is alleged to be negligent or otherwise responsible for any injuries to persons or property.  Such indemnity shall not extend to any claims, demands, damages, judgments, fees (including reasonable attorneys fees), expenses, or other costs to the extent that the same are determined to be the result of the willful misconduct of ST. JUDE, the American Lebanese Syrian Associated Charities, Inc., their present and former trustees, directors, governors, officers, agents, faculty, employees or students Practice of the Patent Rights or sale of Licensed Products by an Affiliate of AMT or an agent or a Sublicensee or a third party on behalf of or for the account of AMT or by a third party who purchases Licensed Product(s) from AMT, shall be considered AMT’s practice of said Patent Rights for purposes of this Section.  The obligation of AMT to defend, indemnify and hold harmless as set out in this Section shall survive the termination of this Agreement, shall continue even after assignment of rights and responsibilities to an Affiliate or Sublicensee, and shall not be limited by any other limitation of liability elsewhere in this Agreement.

 

(c)                                   In the event that it is ultimately determined that AMT is not obligated to indemnify, defend and hold harmless the Indemnitees as against any claims, demands, damages, judgments, fees (including reasonable attorneys fees),

 



 

expenses, or other costs, the Indemnitees shall reimburse AMT for any and all costs and expenses (including lawyers’ fees) incurred by AMT in its defense with respect to the Indemnitees.

 

(ii)                                   Procedures.  The Indemnitees agree to provide AMT with prompt written notice of any claim, suit, action, demand, or judgment for which indemnification is sought under this Agreement.  In no event shall AMT be liable for any claims, demands, damages, judgments, fees (including reasonable attorney’s fees), expenses, or other costs that result from a delay by the Indemnitees in providing AMT with such notice.  AMT agrees, at its own expense, to provide attorneys reasonably acceptable to Indemnitees to defend against any such claim, unless Indemnitees elect to be separately represented (which shall be at Indemnitee’s discretion), in which event any costs incurred by the Indemnitees in relation to retaining their own attorneys shall be the sole responsibility of the Indemnitees.  The Indemnitees shall cooperate fully with AMT in such defense and will permit AMT to conduct and control such defense and the disposition of such claim, suit, or action (including all decisions relative to litigation, appeal, and settlement).  AMT agrees to keep ST. JUDE informed of the progress in the defense and disposition of such claim and to consult with ST. JUDE with regard to any proposed settlement.

 

(iii)                                Insurance.  Prior to initial human testing or sale of any Licensed Products and thereafter so long as Licensed Products are being sold in any particular country AMT shall establish and maintain appropriate insurance coverage in the minimum amount of five million dollars ($5,000,000) per claim, with an aggregate of ten million dollars ($10,000,000), to cover any liability arising from AMT’s indemnification obligations under this Section 8 with respect to such human testing or sale of Licensed Product.  Prior to initial human testing or sale of any Licensed Product and thereafter so long as Licensed Products are being sold in any particular

 



 

country, AMT shall establish and maintain, in each country in which AMT or Sublicensees shall test or sell Licensed Products, product liability or other appropriate insurance coverage in the minimum amount of five million dollars ($5,000,000) per claim.  AMT will annually present evidence, in the form of a statement in the annual diligence report to ST. JUDE that such coverage is being maintained.  Upon ST. JUDE’S request, AMT will furnish ST. JUDE with a Certificate of Insurance of each insurance policy obtained.  ST. JUDE and ALSAC shall be listed as additional insured’s in AMT’s said insurance policies.  If such insurance is underwritten on a ‘claims made’ basis, AMT agrees that any change in underwriters during the term of this Agreement and thereafter so long as Licensed Products are being sold will require the purchase of ‘prior acts’ coverage to ensure that coverage will be continuous throughout the term of this Agreement and thereafter so long as Licensed Products are being sold.

 

9.                                       TERM AND TERMINATION

 

9.1                                Term.  This Agreement shall commence on the Effective Date and, and unless sooner terminated in accordance with any of the provisions herein, expire when no further payment is due from AMT to ST. JUDE hereunder in relation to sales of Licensed Product.

 

9.2                                Voluntary Termination by AMT.  AMT shall have the right to terminate this Agreement, for any reason, upon ninety (90) days, prior written notice to ST. JUDE.  Upon termination, a final report as described in Section 5.2 shall be submitted and any previously arising (before the effective termination date) milestone payments, annual fees, royalty payments, and unreimbursed patent expenses due to ST. JUDE shall become immediately payable.

 

9.3                                Termination for Default.  In the event that either Party commits a material breach of its obligations under this Agreement and fails to cure that breach within sixty (60) days after receiving written notice thereof, the other Party may terminate this Agreement immediately

 



 

upon written notice to the party in breach unless the Party allegedly in breach disputes that a material breach has occurred and submits notice of such dispute to the other Party, following which the Parties shall first try to resolve the dispute within thirty (30) days of such notice and if such dispute cannot be resolved, the dispute shall be subject to the jurisdiction of the courts pursuant to Section 10.7.  For the avoidance of doubt, there shall be no termination of this Agreement pending the outcome of dispute resolution.

 

If the alleged breach involves non payment of any undisputed amounts due ST. JUDE under this Agreement, AMT shall pay an interest penalty based on the amount owed at a daily accrual rate equal to the lesser of ten percent (10%) per annum or the highest rate permissible by law on the unpaid balance until the undisputed amount is paid in full.

 

9.4                                Termination for Insolvency.  ST. JUDE shall have the right to terminate this Agreement on written notice to AMT in the event of the occurrence of insolvency of AMT.

 

9.5                                Effect of Termination on Sublicensees.  If termination under Sections 9.3 or 9.4 of this Agreement is no fault of a Sublicensee ST. JUDE agrees to enter into a direct license of Patent Rights with any Sublicensee on substantially the same terms as the sublicense between AMT and the Sublicensee with respect to terms pertaining to the Patent Rights, provided that the terms of the sublicense are at least as favorable to ST. JUDE as the terms of this Agreement and prior to the making of any such sub-license by AMT, ST. JUDE will undertake directly to such Sublicensee that ST. JUDE will do this.

 

9.6                                Effect of Termination.  Upon termination, AMT shall cease to utilize Patent Rights and shall so certify to ST. JUDE in writing that Patent Rights are not being used for any purpose by AMT.  Termination shall not affect any rights or obligations which have accrued prior to termination or which by their nature are intended to survive termination such as Section 1, 5.2 (obligation to provide final report and payment), 7, 8.1, 9.5, 10.1, 10.7 and 10.8.  Upon the early termination of this Agreement, AMT may complete and sell any work-in-progress

 



 

and inventory of Licensed Products that exists as of the effective date of termination, provided that (i) AMT is current in payment of all amounts due ST. JUDE under this Agreement, (ii) AMT pays ST. JUDE the applicable royalty on such sales of Licensed Products in accordance with the terms and conditions of this Agreement, and (iii) AMT shall complete and sell all work-in-progress and inventory of Licensed Products within six (6) months after the effective date of termination.

 

10.                                MISCELLANEOUS

 

10.1                         Representation and Warranties of both Parties.

 

(i)                                      Each Party hereby represents and warrants to the other Party, as of the Effective Date, as follows:

 

(a)                                  Such Party has the power and authority and legal right to enter into this Agreement, to perform its obligations and to grant the licenses hereunder, and has taken all necessary action on its part to authorize the execution and delivery of this Agreement and the performance of its obligations hereunder;

 

(b)                                  This Agreement has been duly executed and delivered on behalf of such Party and constitutes a legal and valid obligation binding upon such Party and enforceable against it in accordance with its terms;

 

(c)                                   The execution, delivery and performance of this Agreement by such Party do not conflict with any agreement, instrument or understanding, oral or written, to which it is a party or by which it is bound, nor violate any applicable law or regulation of any governmental body or administrative or other agency having jurisdiction over it;

 



 

(d)                                  Such Party is aware of no action, suit, inquiry or investigation instituted by any third party that questions or threatens the validity of this Agreement; and

 

(e)                                   All necessary consents, approvals and authorizations of all governmental authorities and other persons required to be obtained by such Party in connection with this Agreement have been obtained.

 

(ii)                                   Further Representations and Warranties of ST. JUDE:

 

(a)                                  ST. JUDE either legally or beneficially owns or controls the entire right, title and interest in and to the Patent Rights licensed hereunder.

 

(b)                                  there is, to its knowledge as of the Effective Date, no action, suit, claim, proceeding or governmental investigation pending or threatened against ST. JUDE with respect to enforceability of the Patent Rights licensed hereunder, either at law or in equity, before any court or administrative agency or before any governmental department, commission, board, bureau, agency or instrumentality, whether United States or foreign.

 

(c)                                   ST. JUDE has informed AMT in writing of all intellectual property rights of third parties in the Field of which ST. JUDE is aware to the best of ST. JUDE’s knowledge.

 

ST. JUDE MAKES NO OTHER WARRANTIES CONCERNING THE PATENT RIGHTS, INCLUDING WITHOUT LIMITATION ANY EXPRESS OR IMPLIED WARRANTY OF MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE.  Specifically, ST. JUDE makes no warranty or representation (i) regarding the validity or scope of the Patent Rights, (ii) that exploitation of the Patent Rights or any Licensed Product will not infringe any patents or other intellectual property rights of a third

 



 

party, and (iii) that any third party is not currently infringing or will not infringe the Patent Rights.

 

10.2                         Force Majeure.  Neither Party will be responsible for its inability to perform any of its obligations under this Agreement resulting from causes beyond the reasonable control of such Party, including without limitation fires, explosion, flood, war, strike, or riot, provided that the nonperforming Party uses reasonable efforts to avoid or remove such causes of nonperformance and continues performance under this Agreement with reasonable dispatch whenever such causes are removed.

 

10.3                         Headings.  All headings are for convenience only and shall not affect the meaning of any provision of this Agreement.

 

10.4                         Binding Effect.  This Agreement shall be binding upon and inure to the benefit of the parties and their respective permitted successors and assigns.

 

10.5                         Assignment.  The benefit and/or burden of this Agreement may not be assigned by either Party without the prior written consent of the other Party, such consent not to be unreasonably withheld, except that AMT may, without the consent of ST. JUDE assign this Agreement to an Affiliate or to a successor in connection with a merger, consolidation, or sale of all or substantially all of its assets or that portion of its business to which this Agreement relates, but shall notify ST. JUDE of such an assignment within sixty (60) days of its occurrence.  Any assignment in violation of this provision shall be null and void.

 

10.6                         Amendment and Waiver.  This Agreement may be amended, supplemented, or otherwise modified only by means of a written instrument signed by both Parties.  Any waiver of any rights or failure to act in a specific instance shall relate only to such instance and shall not be construed as an agreement to waive any rights or fail to act in any other instance, whether or not similar.

 



 

10.7                         Governing Law.  This Agreement shall be governed by and construed in accordance with the laws of the State of New York irrespective of any conflicts of law principles or choice of law rules of any state or country.  Any lawsuit that may be brought with respect to this Agreement shall be brought and tried in a court of competent jurisdiction in New York.  AMT represents that choice of law provisions agreed to by parties to a written contract are generally honored under Dutch law.

 

10.8                         Notice.  Any notices required or permitted under this Agreement shall be in writing, shall specifically refer to this Agreement, and shall be sent by hand, recognized national overnight courier, confirmed facsimile transmission, or registered or certified mail, postage prepaid, return receipt requested, to the following address or facsimile numbers of the parties:

 

To ST. JUDE:                                                                   Office of Technology Licensing
Mail Stop 0742
St. Jude Children’s Research Hospital
332 North Lauderdale
Memphis, Tennessee 38105  Attn: Director

 

Facsimile: (901) 495-3148

 

To AMT:                                                                                          Amsterdam Molecular Therapeutics B.V.
Meibergdreef 611105 BA
Amsterdam, The Netherlands
Attn: Anthony Gringeri

 

Facsimile: +31 20 566 9272

 

All notices under this Agreement shall be deemed effective upon receipt.  A Party may change its contact information upon written notice to the other Party.

 

10.9                         Severability.  In the event that any provision of this Agreement shall be held invalid or unenforceable for any reason, such invalidity or unenforceability shall not affect any other

 



 

provision of this Agreement, and the Parties shall negotiate in good faith to modify the Agreement to preserve (to the extent possible) their original intent.  During such negotiation, this Agreement shall be construed as if such provision were deleted by agreement of the Parties.

 

10.10                  Entire Agreement.  This Agreement, together with the Sponsored Research Agreement and the AMT Technology License Agreement between the Parties executed concurrently herewith, constitutes the entire agreement between the Parties with respect to the subject matter and supersedes all prior agreements or understandings between the Parties relating to its subject matter.

 

IN WITNESS WHEREOF, the Parties have caused this Agreement to be executed by their duly authorized representatives.

 

 

Amsterdam Molecular Therapeutics B.V.

 

St. Jude Children’s Research Hospital, Inc.

 

 

 

 

 

 

 

 

 

 

By:

/s/ A.J. Gringeri

 

By:

/s/ J. Scott Elmer

 

Anthony Gringeri

 

 

J. Scott Elmer

 

Chief Operating Officer

 

 

Director, Technology Licensing

 

 

 

 

 

Date:

4 July 2008

 

Date:

07/07/08

 



 


 

 

ST. JUDE CHILDREN’S RESEARCH HOSPITAL, INC.

(1)

 

 

 

 

and

 

 

 

 

 

UNIQURE BIOPHARMA BV

(2)

 


 

AMENDMENT N°1 TO THE
EXCLUSIVE LICENSE AGREEMENT

 


 



 

THIS AMENDMENT N°1 TO THE EXCLUSIVE LICENSE AGREEMENT (this “Amendment”), with the effective date of July 12, 2012 (“Effective Date”),

 

BY AND BETWEEN

 

(1)                                  ST. JUDE CHILDREN’S RESEARCH HOSPITAL, INC. , a Tennessee not-for-profit corporation located at 262 Danny Thomas Place, Memphis, Tennessee 38105 (“St. Jude”); and

 

(2)                                  UNIQURE BIOPHARMA BV (formerly: Amsterdam Molecular Therapeutics (AMT) B.V.), a company incorporated under the laws of the Netherlands, with offices at Meibergdreef 61, 1105 BA Amsterdam, The Netherlands (“uniQure”).

 

(each, a “Party” and together the “Parties”)

 

BACKGROUND :

 

(A)                                The Parties have signed an Exclusive License Agreement dated July 7th, 2008 (hereinafter the “Agreement”).

 

(B)                                The Parties desire that the Agreement be amended as set forth below in order to:

 

I.                                         change the name of the licensee from Amsterdam Molecular Therapeutics B.V. (“AMT”) to uniQure biopharma B.V. This name change is the result of a transaction that took place on 30 March 2012, whereby Amsterdam Molecular Therapeutics N.V., a public company, was liquidated and all its operations and stocks were transferred to UniQure B.V., a privately held company;

 

II.                                    add language related to financial terms associated with sublicensing, so as to clarifyfinancial obligations due to St.Jude from sublicensing of the patent rights granted in the Agreement by uniQure in order to expedite the development of therapeutics for rare diseases.

 

IT IS NOW AGREED AS FOLLOWS:

 

1.                                       Modifications

 

I.                                         In the Agreement, all references to “Amsterdam Molecular Therapeutics B.V.” are changed to “uniQure biopharma B.V.”.

 

II.                                    In the Agreement, all references to “AMT” are changed to “uniQure”.

 

III.                               Section 4.5 (i)* is amended to read as follows:

 


* 2 nd  subsection (f) of Section 4.5.

 



 

(i)                                      Support for research, Development and/or manufacturing activities corresponding directly to the Development and commercial manufacture of Licensed Products, which do not exceed the fully-burdened cost for undertaking such research, Development, and/or manufacturing performed by or for AMT (including third parties on AMT’s behalf), each pursuant to a specific agreement including a performance plan and commensurate budget;

 

IV.                                The following Section 4.7 is added to the Agreement:

 

4.7                                Sublicense consideration apportionment. The percentages referred to under subsections (i), (ii) and (iii) immediately below the first paragraph of Section 4.5 shall apply only to that portion of sublicense consideration attributable to sublicensing of the Patent Rights.  In any agreement which includes the grant of a sublicense to Patent Rights along with other rights and assets held by uniQure that are necessary or desirable for the development, manufacture and sale of Licensed Products, the Parties shall agree on the portion of income from such an agreement that should be attributable to sublicensing of the Patent Rights, taking into account the value of the Patent Rights in comparison to the value of the other rights and assets transferred by uniQure to the sublicensee that are necessary or desirable for the development, manufacture and sale of Licensed Products.

 

2.                                       Miscellaneous: All the other provisions of the Agreement remain unchanged and fully applicable between the Parties, and the terms and definitions used in the Agreement shall, so far as possible, apply to this Amendment.

 

 

IN WITNESS WHEREOF , the Parties have caused this Agreement to be executed as of the Effective Date.

 

 

St. Jude Children’s Research Hospital, Inc.

 

UniQure biopharma B.V.

 

 

 

 

 

 

 

 

By:

 

 

By:

/s/ PJ Morgan

Name:

 

 

Name:

PJ Morgan

Title:

 

 

Title:

CFO

Date:

 

 

Date:

12 July 2012

 


Exhibit 10.4

 

CO-DEVELOPMENT AND LICENSE AGREEMENT

 

This Co-Development and License Agreement (this “Agreement”) is entered into as of 29 April 2013 (the “Effective Date”), by and between uniQure Biopharma B.V., formerly known as Amsterdam Molecular Therapeutics (AMT) B.V., a Dutch corporation, with its offices at Meibergdreef 61, 1105 BA Amsterdam, The Netherlands (“uniQure”), and Chiesi Farmaceutici S.p.A., an Italian corporation, with its offices at Via Palermo, 26/A, 43122 Parma, Italy (“Chiesi”). uniQure and Chiesi are sometimes referred to herein individually as a “Party” and collectively as the “Parties”.

 

WHEREAS, uniQure is a company engaged in the research and clinical development of human gene based therapies and uniQure Controls (as defined below) all relevant rights to a certain Gene Therapy Product for Hemophilia B, as more specifically described below;

 

WHEREAS, Chiesi is a pharmaceutical company engaged in the research, development, manufacture and commercialization of products for human diseases and disorders;

 

WHEREAS, Chiesi and uniQure are interested in collaborating in the co-development and future Commercialization (as defined below) of the Product, on the terms and conditions set forth herein.

 

NOW, THEREFORE, uniQure and Chiesi hereby agree as follows:

 

ARTICLE I
DEFINITIONS; INTERPRETATION

 

Capitalized terms used herein shall have the meanings assigned to them as follows.

 

Section 1.1            “AAV5 Vector” shall mean a recombinant adeno-associated virus vector with the serotype 5 that is a non-pathogenic, replication defective, parvovirus engineered to deliver functional gene copies to humans.

 

Section 1.2            “Acquired Party” has the meaning set forth in Section 15.1.

 

Section 1.3            “Acquirer” has the meaning set forth in Section 15.1.

 

Section 1.4            “Additional Rights” has the meaning set forth in Section 7.6(a).

 

Section 1.5            “Additional Rights Agreement” shall mean a written agreement to which either or both Parties are a party and that conveys rights in Additional Rights that are included in Licensed Technology or the Chiesi Technology pursuant to Section 7.6(d).

 

Section 1.6            “Affiliate” shall mean, with respect to a Party, any Person Controlled by, in Control of, or under common Control with such Party.

 

Section 1.7            “Agreement” has the meaning set forth in the first and opening paragraph of this Agreement.

 

1



 

Section 1.8            “Alliance Manager” has the meaning set forth in Section 2.2(f).

 

Section 1.9            “Applicable Laws” shall mean all applicable laws, rules and regulations, including any rules, regulations, guidelines or other requirements of any Regulatory Authority, that may be in effect from time to time.

 

Section 1.10          “Approved Activities” shall mean those activities identified on Exhibit C.

 

Section 1.11          “Arbitration Request” has the meaning set forth in Section 13.2(b)(i).

 

Section 1.12          “Average Net Sales Price” shall mean the average net sales price of a particular Product in the Territory, calculated on a monthly basis, by dividing the Net Sales of the Product in the Territory effected in a particular calendar month by the number of patient doses of the Product accounting for the Net Sales in such calendar month.

 

Section 1.13          “Breaching Party” has the meaning set forth in Section 12.3.

 

Section 1.14          “Business Day” shall mean a day on which banking institutions in Amsterdam, The Netherlands and Parma, Italy, are open for business, excluding any Saturday or Sunday.

 

Section 1.15          “Change of Control” shall mean, with respect to a Party, the acquisition by any Third Party, directly or indirectly, of the Control of such Party.

 

Section 1.16          “Chiesi” has the meaning set forth in the first and opening paragraph of this Agreement.

 

Section 1.17          “Chiesi Know-How” shall mean all Know-How Controlled by Chiesi as of the Effective Date or during the Term that is necessary or useful for the Development, use, Manufacture, having Manufactured, or Commercialization of the Product. Chiesi Know-How also includes any Joint Know-How to the extent Controlled by Chiesi, and such other Know-How included in the Chiesi Know-How pursuant to Section 7.6.

 

Section 1.18          “Chiesi Patents” shall mean all Patent Rights Controlled by Chiesi as of the Effective Date or during the Term that are directed to any Chiesi Know-How or the Development, Manufacture, having Manufactured, use, or Commercialization of the Product. Chiesi Patents also include any Joint Patents to the extent Controlled by Chiesi, and such other Patent Rights included in the Chiesi Patents pursuant to Section 7.6.

 

Section 1.19          “Chiesi Shared Costs” has the meaning set forth in Section 8.2(a).

 

Section 1.20          “Chiesi Sole Costs” shall mean all costs incurred in connection with launch and pre-launch commercial activities for the Product in the Territory, including key opinion leader (KOL) development, market research, pricing and reimbursement studies.

 

2



 

Section 1.21          “Chiesi Technology” shall mean Chiesi Know-How and Chiesi Patents.

 

Section 1.22          “Claims” has the meaning set forth in Section 14.1.

 

Section 1.23          “CMC” shall mean the Chemistry, Manufacturing and Controls portion of any IMPD or Marketing Authorization Application.

 

Section 1.24          “Collaboration” shall mean the relationship between and activities conducted by the Parties under this Agreement and all other agreements between the Parties referenced herein (other than the Confidentiality Agreement), including the Commercialization Agreement (collectively, the “Collaboration Agreements”).

 

Section 1.25          “Collaboration Agreements” has the meaning set forth in Section 1.24.

 

Section 1.26          “Commercialization Agreement” shall mean that certain Commercialization Agreement for Glybera concluded separately between the Parties on the date hereof.

 

Section 1.27          “Commercialization” shall mean any and all activities, whether before or after Regulatory Approval, directed to the marketing, detailing and promotion of the Product and shall include pre-launch, launch and post-launch marketing, promoting, detailing, marketing research, medical affairs, managed markets, distributing, offering to commercially sell and commercially selling the Product, importing, exporting or transporting the Product for commercial sale and regulatory affairs with respect to the foregoing, including the filing and obtaining of Price and Reimbursement Approval for the Product, but shall not include Manufacturing nor any Development activities. When used as a verb, “Commercializing”, “Commercialize” and “Commercialized” shall mean to engage in Commercialization.

 

Section 1.28          “Commercially Reasonable Efforts” shall mean, with respect to the efforts to be expended by a Party with respect to a goal, reasonable, diligent, good faith efforts to accomplish such goal as a similarly situated (with respect to size, stage of development, and assets) biotechnology or pharmaceutical company, as the case may be, would use to accomplish a similar goal under similar circumstances so as to achieve such goal as expeditiously as possible; provided that, with respect to the Development and Commercialization of the Product, such efforts shall be substantially equivalent to those efforts and resources that a similarly situated (with respect to size, stage of development, and assets) biotechnology or pharmaceutical company, as the case may be, would typically devote to its own internally discovered products of similar market potential at a similar stage in their development or product life so as to achieve such goal as expeditiously as possible (which, with respect to activities for which Chiesi is responsible, shall be without regard to any amounts paid or payable to uniQure with respect to the Product under this Agreement, the Commercialization Agreement, or the HemB Supply and Distribution Agreement).

 

3



 

Section 1.29          “Competing Product” shall mean any Gene Therapy product developed, manufactured or commercialized in the Field other than the Product Developed, Manufactured or Commercialized pursuant to this Agreement.

 

Section 1.30          “Competitive Infringement” has the meaning set forth in Section 9.3(a).

 

Section 1.31          “Confidential Information” shall mean all confidential or proprietary information of a Party, including information regarding such Party’s or its Affiliates’ or licensors’ technology, products, business, business plans, financial status, biological substances, chemical substances, formulations, techniques, methodology, equipment, sources of supply and patent positioning and information belonging to such Party’s Affiliate or a Third Party provided to the other Party under this Agreement. The status, prospects or objectives regarding the Development Program or any Product being developed hereunder, as well as the terms and conditions of this Agreement, shall be deemed “Confidential Information” of both Parties. All information disclosed by uniQure prior to the Effective Date pursuant to the Two Way Confidentiality Disclosure Agreement between Amsterdam Molecular Therapeutics (AMT) B.V. and Chiesi Farmaceutici S.p.A., dated 22 July 2010 (the “Confidentiality Agreement”) shall be deemed “Confidential Information” of uniQure hereunder.

 

Section 1.32          “Confidentiality Agreement” has the meaning set forth in Section 1.31.

 

Section 1.33          “Control” or “Controlled” shall mean (a) when used in reference to any Confidential Information, Know-How, Patent or other intellectual property rights, the possession (whether by ownership or license (other than solely pursuant to a license under this Agreement)) by such Party or any of its Affiliates, of the legal authority or right to grant to the other Party access or a license or sublicense to such Confidential Information, Know-How, Patent or other intellectual property rights as provided herein, without violating the terms of any agreement or arrangement with any Third Party, or (b) when used in reference to Section 1.6, Section 1.15 and Section 15.1, (i) the possession, directly or indirectly, of the power to direct the management or policies of a Person, whether through ownership of voting securities, by contract or otherwise; (ii) ownership of fifty percent (50%) or more of the voting securities entitled to vote for the election of directors in the case of a corporation, or fifty percent (50%) or more of the equity interest in the case of any other type of legal entity; or (iii) status as a general partner in any partnership, or any other arrangement whereby a Person controls or have the right to control the board of directors or equivalent governing body of a corporation or other Person. Notwithstanding the foregoing, any portfolio company of any stockholder of such Person (which stockholder is a venture capital fund or private equity fund) shall not be deemed to be “under common Control with” such Person.

 

Section 1.34          “Controlling Party” has the meaning set forth in Section 7.6(b).

 

Section 1.35          “Cover” or “Covered” shall mean, with respect to any Patent and the subject matter at issue, that, but for a license granted under a Valid Claim of such Patent, the manufacture, use, sale, offer for sale or importation of the subject matter at issue would infringe such Valid Claim, or, in the case of a Patent that is a

 

4



 

patent application, would infringe a Valid Claim in such patent application if it were to issue as a patent.

 

Section 1.36          “Development” shall mean all non-clinical and clinical drug development activities, each to the extent reasonably relating to the development of the Product in the Territory.  Development shall include toxicology, pharmacology, and other non-clinical efforts, test method development and stability testing, formulation development, delivery system development, quality assurance and quality control development, statistical analysis, the conduct of clinical trials or other activities, including regulatory activities, relating to obtaining Regulatory Approval.  When used as a verb, “Develop” means to engage in Development activities.

 

Section 1.37          “Development Plan and Budget” shall mean a plan and budget setting forth the Development activities with respect to the Product and the budget therefor, as prepared, updated and amended from time to time in accordance with this Agreement. The Development Plan and Budget includes the Initial Development Plan and Budget.

 

Section 1.38          “Development Program” shall mean the pre-clinical, CMC, clinical and other development, regulatory and other pre-Marketing Authorization commercial activities of the Parties directed to the Product and undertaken in accordance with the Development Plan and Budget.

 

Section 1.39          “Effective Date” has the meaning set forth in the first and opening paragraph of this Agreement.

 

Section 1.40          “EMA” shall mean the European Medicines Agency and any successor agency thereto.

 

Section 1.41          “EPO” has the meaning set forth in Section 9.2(d)(iii).

 

Section 1.42          “EU” shall mean the European Union.

 

Section 1.43          “EU Member States” shall mean Austria, Belgium, Bulgaria, Cyprus, the Czech Republic, Denmark, Estonia, Finland, France, Germany, Greece, Hungary, Ireland, Italy, Latvia, Lithuania, Luxembourg, Malta, the Netherlands, Poland, Portugal, Romania, Slovakia, Slovenia, Spain, Sweden and the United Kingdom.

 

Section 1.44          “Executive Officers” shall mean the Chief Executive Officer of Chiesi or a senior officer designated by Chiesi, and the Chief Executive Officer of uniQure or a senior officer designated by uniQure.

 

Section 1.45          “Existing Third Party Licenses” shall mean the PHS Agreements, the PSC Agreement and the St. Jude Agreements.

 

Section 1.46          “FDA” shall mean the US Food and Drug Administration and any successor agency thereto.

 

Section 1.47          “Field” shall mean the treatment of Hemophilia B in humans.

 

5



 

Section 1.48          “First Commercial Sale” shall mean, the first sale by Chiesi, an Affiliate of Chiesi, or a Sub-distributor of Chiesi, as the case may be, of the Product to a Third Party in the Territory; provided, however, that neither (a) transfers of the Product between Chiesi and its Affiliates, or Sub-distributors nor (b) supply of the Product for clinical trial purposes, shall constitute a First Commercial Sale.

 

Section 1.49          “FTE” shall mean a full time equivalent person year (consisting of a total of one thousand six hundred (1,600) hours per year) of work on or directly related to activities undertaken by uniQure hereunder and related management activities.

 

Section 1.50          “FTE Rate” shall mean EUR 250,000 per FTE, which includes overhead expenses and bench fees (materials used during Development, but excluding expenses for materials and external costs for GMP manufacturing).

 

Section 1.51          “Fully Loaded Cost of Goods” shall mean the fully loaded cost of goods of the Product as defined in Exhibit D.

 

Section 1.52          “Gene Therapy” shall mean the introduction and expression of genetic material in cells of a person in order to cure a disease or to minimize disease symptoms.

 

Section 1.53          “HemB Supply and Distribution Agreement” has the meaning set forth in Section 3.4.

 

Section 1.54          “HHS” has the meaning set forth in Section 1.91.

 

Section 1.55          “ICC” shall mean the International Chamber of Commerce.

 

Section 1.56          “IMPD” shall mean an application submitted to a Regulatory Authority to initiate human clinical trials, including (a) an Investigational Medicinal Product Dossier required to be submitted to the EMA or other Regulatory Authorities in the EU Member States for Regulatory Approval of clinical trials in the EU Member States, as further defined in the Clinical Trials Directive (2001/20/EC), (b) any non-EU Member States equivalent of the foregoing in any other country, and (c) all supplements and amendments that may be filed with respect to the foregoing.

 

Section 1.57          “Improvement” shall mean any invention, discovery, development or modification relating to the Licensed Technology or the Product or the development, manufacture or exploitation thereof, including any method or process of manufacturing or using the Product, and any formulation for the Product, whether or not patentable as well as packaging and labeling of the Product, in each case if and to the extent Controlled by uniQure during the Term.

 

Section 1.58          “In-Person JDC Meetings” has the meaning set forth in Section 2.2(c)(i).

 

Section 1.59          “Indemnified Party” has the meaning set forth in Section 14.3(a).

 

6



 

Section 1.60          “Indemnifying Party” has the meaning set forth in Section 14.3(a).

 

Section 1.61          “Initial Development Plan and Budget” shall mean the activities and budget set forth on Exhibit A attached hereto.

 

Section 1.62          “Invalidity Claim” has the meaning set forth in Section 9.5.

 

Section 1.63          “JCC” has the meaning set forth in Section 2.3(a).

 

Section 1.64          “JDC” has the meaning set forth in Section 2.2(a).

 

Section 1.65          “Joint Know-How” has the meaning set forth in Section 9.1(c).

 

Section 1.66          “Joint Patents” shall mean all Patent Rights that are directed to any Joint Know-How.

 

Section 1.67          “JSC” has the meaning set forth in Section 2.1(a).

 

Section 1.68          “Know-How” shall mean all technical, scientific, manufacturing, financial, commercial and other know-how, data, tangible materials, information, trade secrets, ideas, formulae, inventions, discoveries, processes, machines, compositions of matter, formulations, improvements, protocols, techniques, works of authorship, and results of experimentation and testing (whether or not patentable) in written, electronic, physical (including in the form of tangible compounds or cell lines), oral or any other form.

 

Section 1.69          “License” has the meaning set forth in Section 7.1.

 

Section 1.70          “Licensed Know-How” shall mean all Know-How Controlled by uniQure as of the Effective Date or during the Term that is necessary or useful to Develop, use, or Commercialize the Product in the Field in the Territory. Licensed Know-How also includes any Joint Know-How to the extent Controlled by uniQure, and such other Know-How included in the Licensed Know-How pursuant to Section 7.6(c).

 

Section 1.71          “Licensed Patents” shall mean (a) all Patent Rights Controlled by uniQure as of the Effective Date or during the Term that are necessary or useful to Develop, use, or Commercialize the Product in the Field in the Territory, including those Patent Rights set forth in Exhibit E, which exhibit shall be updated or confirmed upon the date this Agreement has become effective pursuant to Section 12.1(b); (b) any Joint Patents to the extent Controlled by uniQure; and (c) such other Patent Rights included in the Licensed Patents pursuant to Section 7.6(c).

 

Section 1.72          “Licensed Technology” shall mean Licensed Know-How and Licensed Patents.

 

Section 1.73          “Losses” has the meaning set forth in Section 14.1.

 

Section 1.74          “Major Country” shall mean any of the following countries: France, Germany, Italy, Spain and the United Kingdom.

 

7



 

Section 1.75          “Manufacture” and “Manufacturing” shall mean all activities related to the production, manufacture, processing, filling, finishing, packaging, labeling, shipping and holding of the Product or any intermediate thereof, including process development, process qualification and validation, scale up, pre-clinical, clinical and commercial manufacture and analytical development, product characterization, stability testing, quality assurance and quality control. When used as a verb, “Manufacture” shall mean to engage in Manufacturing activities.

 

Section 1.76          “Marketing Authorization” or “MA” shall mean the authorization issued by the relevant Regulatory Authority necessary to place on the market the Product in any country or regulatory jurisdiction in the Territory (including the approval of a Marketing Authorization Application in the EU Member States). For clarity, a Marketing Authorization shall not include any applicable Price and Reimbursement Approvals.

 

Section 1.77          “Marketing Authorization Application” or “MAA” shall mean an application submitted to a Regulatory Authority for marketing approval of a drug or biologic product, including (a) a Marketing Authorization Application in the EU Member States under Regulation (EC) No. 726/2004 or Directive 2001/83/EC, (b) any non-EU Member States equivalent of the foregoing in any other country in the Territory, and (c) all supplements and amendments that may be filed with respect to any of the foregoing.

 

Section 1.78          “Net Sales” shall mean the total amount of invoiced sales of the Product in the Territory by or on behalf of Chiesi or its Affiliates or Sub-distributors to Third Parties (including wholesalers, hospitals, end users and others), in bona fide arm’s length transactions, less the following deductions, in each case related specifically to the Product and customary in the trade and actually allowed and taken by such Third Parties and not otherwise recovered by or reimbursed to Chiesi: (a) cash discounts; (b) taxes on sales (such as sales or use taxes) to the extent added to the sale price and set forth separately as such in the total amount invoiced; (c) freight and insurance to the extent added to the sale price and set forth separately as such in the total amount invoiced; (d) amounts repaid or credited by reason of rejections, defects, recalls, expirations, or returns; and (e) any governmental mandated charge backs, rebates, and discounts. No deductions shall be made for (x) commissions paid to individuals, whether they are with independent sales agencies or regularly employed by Chiesi or any of its Affiliates, and on its payroll, (y) the cost of collections, and (z) any advertising and promotional expenses.

 

Section 1.79          “New Product” has the meaning set forth in Section 6.2(a).

 

Section 1.80          “NIH” has the meaning set forth in Section 1.92.

 

Section 1.81          “Non-Acquired Party” has the meaning set forth in Section 15.1.

 

Section 1.82          “Non-Arbitrable Termination Dispute” has the meaning set forth in Section 13.1(a).

 

Section 1.83          “Non-Breaching Party” has the meaning set forth in Section 12.3.

 

8



 

Section 1.84          “Non-Controlling Party” has the meaning set forth in Section 7.6(b).

 

Section 1.85          “Party” and “Parties” has the meaning set forth in the first and opening paragraph of this Agreement.

 

Section 1.86          “Patent Challenge” has the meaning set forth in Section 12.4.

 

Section 1.87          “Patent Prosecution” shall mean the preparation, filing and prosecution of patent applications, and the maintenance of patents, included in Patent Rights.

 

Section 1.88          “Patent Right(s)” shall mean any patent or patent application, including utility patents, utility models, design patents, provisional applications, certificates of invention, and all divisionals, continuations, continuations-in-part, substitutions, reissues, reexaminations, renewals, extensions (including any supplemental patent certificate) or additions to any patent or patent application.

 

Section 1.89          “Paying Party” has the meaning set forth in Section 8.4.

 

Section 1.90          “Person” shall mean any natural person or any corporation, company, partnership, limited liability company, joint venture, firm, agency or other entity, including a Party.

 

Section 1.91          “Phase I/II Study” shall mean the first human clinical trial in which patients with Hemophilia B are dosed with the Product.

 

Section 1.92          “PHS” shall mean The National Institutes of Health (“NIH”) or the FDA, agencies of the US Public Health Service within the Department of Health and Human Services (“HHS”).

 

Section 1.93          “PHS Agreements” shall mean the PHS 2011 Agreement and the PHS 2007 Agreement.

 

Section 1.94          “PHS 2007 Agreement” shall mean the Non-Exclusive Patent License Agreement, number L-107-2007/0, dated as of 25 April/2 May 2007, by and between uniQure and PHS, as amended from time to time.

 

Section 1.95          “PHS 2011 Agreement” shall mean the Exclusive and Non-Exclusive Patent License Agreement, number L-116-2011/0, dated as of 5/10 August 2011, by and between uniQure and PHS, as amended from time to time.

 

Section 1.96          “Pivotal Study” shall mean, with respect to the Product, a human clinical trial, the principal purpose of which is to establish safety and efficacy of such Product in patients with Hemophilia B as required under Regulation (EC) No. 726/2004 or Directive 2001/83/EC, or a similar clinical trial prescribed by the applicable Regulatory Authority in the Territory. A Pivotal Study also includes any other human clinical trial intended as a pivotal study of such Product regarding Hemophilia B, such as a phase II/III or phase IIb clinical trial, whether or not such study is a traditional phase III clinical trial.

 

9



 

Section 1.97          “Pre-Existing Affiliate” has the meaning set forth in Section 15.1.

 

Section 1.98          “Price and Reimbursement Approval” shall mean any approval or authorization of any Regulatory Authority establishing a pricing- and payment scheme or a reimbursement scheme for the Product in any country or jurisdiction of the Territory.

 

Section 1.99          “Product” shall mean a Gene Therapy product for the treatment of Hemophilia B in humans that includes an AAV5 Vector containing a functional copy of the codon-optimized human Factor IX gene or part thereof under the control of a liver-specific promoter.

 

Section 1.100       “Product Data” shall mean all preclinical and clinical data, safety data and all other supporting data, including pharmacology and biology data, regulatory documentation (including IMPDs, MAAs and other Marketing Authorizations, Regulatory Approvals and regulatory filings in the Territory) and other Know-How generated under the Development Program that relate to the Product.

 

Section 1.101       “Product Manufacturing Cost Reimbursement” has the meaning set forth on Exhibit D.

 

Section 1.102       “Product Transfer Price” has the meaning set forth on Exhibit D.

 

Section 1.103       “PSC” shall mean Protein Sciences Corporation.

 

Section 1.104       “PSC Agreement” shall mean the License Agreement, dated as of 22 March 2007, by and between uniQure and PSC, as amended from time to time.

 

Section 1.105       “Publishing Party” has the meaning set forth in Section 10.5(a).

 

Section 1.106       “Receiving Party” has the meaning set forth in Section 8.4.

 

Section 1.107       “Reconciliation Payment” has the meaning set forth in Section 8.2(d).

 

Section 1.108       “Regular JDC Meeting” has the meaning set forth in Section 2.2(c)(i).

 

Section 1.109       “Regulatory Approval” shall mean any and all approvals (including, where required, any applicable Price and Reimbursement Approvals), licenses, registrations or authorizations of any Regulatory Authority necessary for the Manufacture, use, and Commercialization of a Product in a country or jurisdiction, including IMPDs and Marketing Authorizations.

 

Section 1.110       “Regulatory Authority” shall mean any federal, national, multinational, state, provincial or local regulatory agency, department, bureau or other governmental entity with authority over the testing, Regulatory Approval, manufacture, use, storage, import, promotion, marketing or sale of a drug or biologic product in a country or jurisdiction, including the EMA.

 

10



 

Section 1.111       “SDEA” has the meaning set forth in Section 4.4.

 

Section 1.112       “SEC” has the meaning set forth in Section 10.1(e).

 

Section 1.113       “Severed Clause” has the meaning set forth in Section 15.14.

 

Section 1.114       “Shared Costs” shall mean, for a calendar quarter, (a) each Party’s duly documented out-of-pocket costs incurred during such calendar quarter pursuant to the Development Program, including costs associated with preclinical studies, clinical studies, CRO, CMC, clinical supply and other reasonable and customary development expenses, as set forth in the Development Plan and Budget; (b) uniQure’s FTE Rate for each uniQure FTE conducting activities during such calendar quarter pursuant to the Development Program (including any activities conducted in connection with the Development Plan and Budget during the period from the Effective Date until the date this Agreement has become effective pursuant to Section 12.1(b)); (c) each Party’s out-of-pocket costs and expenses incurred during such calendar quarter associated with Patent Prosecution of Joint Patents in the Territory as provided in Section 9.2 and enforcement of Joint Patents against Competitive Infringement in the Territory as provided in Section 9.3, including (i) out-of-pocket costs incurred in gathering information or making filings with any governmental authority, (ii) fees and expenses of counsel and consultants (including translators) and (iii) extraordinary employee costs; (d) actual costs associated with any Approved Activity, whether incurred before or after the Effective Date, including as set forth in Exhibit C; (e) development and regulatory milestone payments associated with the Existing Third Party Licenses and, if required, any costs associated with any Additional Rights Agreements, to the extent incurred in connection with Development Program activities performed during such calendar quarter; (f) all costs and expenses relating to the withdrawal or recall of any Product in a country in the Territory prior to, Marketing Approval in such country pursuant to Section 4.3 and (g) all costs related to Phase IV (a post approval study) if agreed between the Parties. Shared Costs shall exclude uniQure Sole Costs and Chiesi Sole Costs.

 

Section 1.115       “St. Jude” shall mean St. Jude Children’s Research Hospital, Inc.

 

Section 1.116       “St. Jude Agreements” shall mean the Exclusive License Agreement and the Sponsored Research Agreement, both dated as of 7 July 2008, between St. Jude and uniQure, as amended from time to time.

 

Section 1.117       “Sub-distributor” shall mean a Third Party that is granted a sub-distribution or other Commercialization right in the Territory by Chiesi under this Agreement.

 

Section 1.118       “Subject Disclosure” has the meaning set forth in Section 10.3(b).

 

Section 1.119       “Supply Failure” has the meaning set forth in Section 3.5.

 

Section 1.120       “Term” has the meaning set forth in Section 12.1.

 

11



 

Section 1.121       “Territory” shall mean the EU Member States, Iceland, Liechtenstein and Norway as well as Albania, Algeria, Andorra, Bosnia, Brazil, Croatia, Egypt, Macedonia, Mexico, Monaco, Montenegro, Morocco, Pakistan, Republic of San Marino, Russia and ex-CIS countries ( i.e. Armenia, Azerbaijan, Belarus, Georgia, Kazakhstan, Kirghizstan, Moldova, Tajikistan, Turkmenistan, Ukraine and Uzbekistan), Serbia (including Kosovo), Switzerland, Tunisia, Turkey, and Vatican City. For the avoidance of doubt, the Territory shall exclude China and the US.

 

Section 1.122       “Third Party” shall mean any Person other than uniQure, Chiesi, or their respective Affiliates.

 

Section 1.123       “uniQure” has the meaning set forth in the first and opening paragraph of this Agreement.

 

Section 1.124       “uniQure Shared Costs” has the meaning set forth in Section 8.2(a).

 

Section 1.125       “uniQure Sole Costs” shall mean (a) any expenses for extra patients in a clinical trial of the Product beyond the number required by Regulatory Authority guidelines, or through Regulatory Authority feedback, in the Territory, unless the inclusion of such extra patients is agreed to by the JDC, (b) post-Marketing Authorization Product Manufacturing costs unless specifically required by Chiesi, (c) any filing fees associated with CMC of the Product and (d) subject to Section 4.3, the costs of safety monitoring of the Product, including any filing fees associated with Pharmacovigliance.

 

Section 1.126       “Upfront Payment” has the meaning set forth in Section 8.1.

 

Section 1.127       “US” or “USA” shall mean the United States of America, including its territories and possessions.

 

Section 1.128       “Valid Claim” shall mean any claim within an issued and unexpired Patent or pending Patent application that (i) is not expired, lapsed, or abandoned, (ii) is not dedicated to the public, disclaimed, or admitted to be unenforceable or invalid; and (iii) has not been invalidated, held unenforceable or cancelled by a court or administrative agency of competent jurisdiction in an order or decision from which no appeal has been or can be taken, including through opposition, re-examination, reissue, disclaimer or otherwise.

 

Section 1.129       “WIPO” has the meaning set forth in Section 9.2(d)(iii).

 

Section 1.130       Interpretation. Any reference in this Agreement to an Article, Section, subsection, paragraph, clause, or Exhibit shall be deemed to be a reference to any Article, Section, subsection, paragraph, clause, or Exhibit, of or to, as the case may be, this Agreement. Except where the context clearly otherwise requires, (a) wherever used, the use of any gender will be applicable to all genders, (b) the singular shall include the plural and vice versa, (c) any definition of or reference to any agreement, instrument or other document refers to such agreement, instrument other document as from time to time amended, supplemented or otherwise modified (subject to any restrictions on such amendments, supplements or modifications set

 

12



 

forth herein or therein), (d) any reference to any Applicable Laws refers to such Applicable Laws as from time to time enacted, repealed or amended, (e) the words “herein”, “hereof” and “hereunder”, and words of similar import, refer to this Agreement in its entirety and not to any particular provision hereof, (f) the words “include”, “includes” and “including” are deemed to be followed by the phrase “but not limited to”, “without limitation” or words of similar import, (g) the word “or” has the inclusive meaning ( i.e. , “and/or”), (h) the word “day” means a calendar day, the word “month” means a calendar month, and the word “year” means, and the word “annual” refers to, a calendar year, (i) the word “quarterly” refers to a calendar quarter, (j) each accounting term used herein that is not specifically defined herein has the meaning given to it under the International Financial Reporting Standards, and (k) the captions or headings of the Exhibits, Articles, Sections or other subdivisions hereof are inserted only as a matter of convenience or for reference and shall have no effect on the meaning of the provisions hereof.

 

ARTICLE II
GOVERNANCE; DECISION MAKING

 

Section 2.1            Joint Steering Committee.

 

(a)           Formation and Membership. Within thirty (30) days after the Effective Date, Chiesi and uniQure shall establish a joint steering committee (the “JSC”) to manage the Collaboration. The JSC to be established under this Agreement shall be identical to the one to be established under the Commercialization Agreement. The JSC shall be comprised of three (3) executives or senior employees of Chiesi and three (3) executives or senior employees of uniQure with appropriate experience and level of decision-making authority. From time to time, in addition to the JDC described below, the Parties may establish one or more subcommittees of the JSC to oversee particular projects or activities ( e.g. , activities under the Commercialization Agreement, financial reporting). Each such subcommittee shall be comprised of an equal number of representatives from each Party with appropriate experience and level of decision-making authority. Each subcommittee shall meet with a frequency to be agreed on by the Parties. Each Party may change any one or more of its representatives on the JSC or any subcommittee at any time upon written notice to the other Party.

 

(b)           Responsibilities.  The JSC shall be responsible for:

 

(i)        providing overall direction of the Collaboration;

 

(ii)       attempting to resolve disputes arising under the Collaboration Agreements; and

 

(iii)      performing such other tasks and undertaking such other responsibilities as may be set forth in the Collaboration Agreements.

 

(c)           Meetings.

 

(i)        The JSC shall meet at least once each calendar quarter, by tele- or video-conference or in person, with the meetings in approximately April and October to be held in-person. The location of in-person JSC meetings shall alternate

 

13



 

between the headquarters offices of each Party, with the first meeting to take place at uniQure’s site in Amsterdam.

 

(ii)       Each Party shall use reasonable efforts to cause its representatives to attend the meetings of the JSC and any subcommittees. In addition, each Party may, at its discretion, invite a reasonable number of non-voting employees or officers, and, with the consent of the other Party, consultants or scientific advisors, to attend meetings of the JSC or any subcommittee, or the relevant portion thereof; provided that, its representatives and any such other employees, officers, consultants or scientific advisors are bound by written obligations of confidentiality that are at least as stringent as those set forth in this Agreement. Each Party shall bear all travel and living expenses of its representatives and other employees, officers, consultants or scientific advisors incurred to attend the meetings of the JSC or any subcommittee.

 

(iii)      Either Party may also request, by providing written notice to the other Party, that a special meeting of the JSC be convened for the purpose of resolving disputes in connection with, or for the purpose of reviewing or making a decision pertaining to, any material matter within the purview of the JSC, the examination or resolution of which cannot reasonably be postponed until the next scheduled JSC meeting. Such meeting shall be convened at such time as may be mutually agreed upon by the Parties, but in any event shall be held within fifteen (15) days after the date of such notice.

 

(d)           Administrative Matters.  The right to appoint the chairperson of the JSC shall alternate on an annual basis between Chiesi and uniQure, with uniQure having the right to appoint the chairperson for the first year of the Term. The Alliance Managers (defined below) shall work with the chairperson to develop JSC meeting agendas. The chairperson shall be responsible for calling meetings of the JSC and for leading the meetings. A JSC member of the chairing Party shall serve as secretary of such meetings. The secretary shall promptly prepare and distribute to all members of the JSC draft minutes of the meeting for review and comment, including a list of any actions or decisions approved by the JSC, with the goal of distributing final approved minutes of each JSC meeting within thirty (30) days after the meeting. Neither the chairperson nor the secretary shall have any greater authority than any other representative on the JSC and the Party appointing the chairperson and the secretary shall not have any greater authority than the other Party by virtue of its right to make such appointments. The chairperson shall include on the agenda any items proposed by either Party.

 

(e)           Decision Making.  Each Party, through its representatives, shall have one (1) vote on the JSC and each subcommittee. Both Parties must vote in the affirmative to allow the JSC or a subcommittee to take any action that requires the approval of the JSC or the subcommittee. Decision on any matter may be taken at a meeting, by teleconference, videoconference or by written agreement. If a subcommittee is unable to resolve any dispute, or to unanimously agree on any matter, within its responsibilities, such dispute or matter shall be referred to the JSC for resolution. Either Party may convene a special meeting of the JSC in accordance with Section 2.1(c)(iii) for the purpose of resolving any dispute within the JSC’s jurisdiction that represents a material issue the resolution of which cannot reasonably await until the next scheduled meeting of the JSC.

 

14



 

(f)            Dispute Resolution by Executive Officers.

 

(i)        If the JSC is unable to resolve any dispute within the responsibilities of the JSC specified in Section 2.1(b) within thirty (30) days after a Party provides notice to the other Party of the existence of such dispute, such dispute or other matter shall be referred to the Executive Officers for resolution, pursuant to Section 13.2.

 

(ii)       In resolving any disputes under this Section 2.1(f), each Party shall act in good faith, subject to the terms and conditions of the Collaboration Agreements, and in a commercially reasonable manner without favoring other products being developed or commercialized by or on behalf of such Party or its Affiliates outside of the Collaboration.

 

Section 2.2            Joint Development Committee.

 

(a)           Formation and Membership.  Within thirty (30) days after the Effective Date, Chiesi and uniQure shall establish, as a subcommittee of the JSC, a joint development committee (the “JDC”) to manage the overall relationship between the Parties under this Agreement. The JDC shall be comprised of three (3) executives or senior employees of Chiesi and three (3) executives or senior employees of uniQure with appropriate experience and level of decision-making authority. From time to time, the Parties may establish one or more subcommittees of the JDC to oversee particular projects or activities ( e.g. , clinical/regulatory, CMC, development). Each such subcommittee shall be comprised of an equal number of representatives from each Party with appropriate experience and level of decision-making authority. Each subcommittee shall meet with a frequency to be agreed on by the Parties. Each Party may change any one or more of its representatives on the JDC or any subcommittee at any time upon written notice to the other Party.

 

(b)           Responsibilities.  The JDC shall be responsible for:

 

(i)        periodically reviewing the Development Plan and Budget and suggesting or approving such updates or amendments to the Development Plan and Budget as the JDC deems appropriate, including all budgets relating to activities to be conducted hereunder and amendments thereto;

 

(ii)       ensuring consistency and coordination between Development activities to be conducted by uniQure under the Development Plan and Budget and, if applicable, by Chiesi under the Development Plan and Budget;

 

(iii)      providing overall strategic direction with respect to Development and regulatory activities for the Product, including activities conducted under the Development Plan and Budget;

 

(iv)      overseeing Development and regulatory activities for the Product;

 

(v)       discussing and addressing any supply chain or other delivery issues that have arisen or might arise relating to the Product;

 

15



 

(vi)      determining, managing and reviewing the Patent strategy relating to inventions made after the Effective Date covering the Product in the Territory;

 

(vii)     attempting to resolve disputes arising under this Agreement that are referred to the JDC by either Party or any subcommittee; and

 

(viii)    performing such other tasks and undertaking such other responsibilities as may be set forth in this Agreement or as may be delegated to it by the JSC.

 

(c)           Meetings.

 

(i)        The JDC shall meet at least once each calendar quarter, by tele- or video-conference or in person, with the meetings in approximately March and September to be held in-person (such two (2) regularly scheduled March and September in-person meetings shall be the “Regular JDC Meetings”, while all in-person meetings of the JDC, including the Regular JDC Meetings, shall be “In-Person JDC Meetings”). The location of In-Person JDC Meetings shall alternate between the headquarters offices of each Party, with the first meeting to take place at uniQure’s site in Amsterdam.

 

(ii)       Each Party shall use reasonable efforts to cause its representatives to attend the meetings of the JDC and any subcommittees. In addition, each Party may, at its discretion, invite a reasonable number of non-voting employees or officers, and, with the consent of the other Party, consultants or scientific advisors, to attend meetings of the JDC or any subcommittee, or the relevant portion thereof; provided that, its representatives and any such other employees, officers, consultants or scientific advisors are bound by written obligations of confidentiality that are at least as stringent as those set forth in this Agreement. Each Party shall bear all travel and living expenses of its representatives and other employees, officers, consultants or scientific advisors incurred to attend the meetings of the JDC or any subcommittee.

 

(iii)      Either Party may also request, by providing written notice to the other Party, that a special meeting of the JDC be convened for the purpose of resolving disputes in connection with, or for the purpose of reviewing or making a decision pertaining to, any material matter within the purview of the JDC, the examination or resolution of which cannot reasonably be postponed until the next scheduled JDC meeting. Such meeting shall be convened at such time as may be mutually agreed upon by the Parties, but in any event shall be held within fifteen (15) days after the date of such notice.

 

(d)           Administrative Matters.  uniQure shall have the right to appoint the chairperson of the JDC. The Alliance Managers (defined below) shall work with the chairperson to develop JDC meeting agendas. The chairperson shall be responsible for calling meetings of the JDC and for leading the meetings. A uniQure JDC member shall serve as secretary of such meetings. The secretary shall promptly prepare and distribute to all members of the JDC draft minutes of the meeting for review and comment, including a list of any actions or decisions approved by the JDC, with the goal of distributing final approved minutes of each JDC meeting within thirty (30) days after the meeting. Neither the chairperson nor the secretary shall have

 

16



 

any greater authority than any other representative on the JDC and uniQure shall not have any greater authority than Chiesi by virtue of its right to make such appointments. The chairperson shall include on the agenda any items proposed by either Party.

 

(e)           Decision Making.  Each Party, through its representatives, shall have one (1) vote on the JDC and each subcommittee. Both Parties must vote in the affirmative to allow the JDC or a subcommittee to take any action that requires the approval of the JDC or the subcommittee. Decision on any matter may be taken at a meeting, by teleconference, videoconference or by written agreement. If a subcommittee is unable to resolve any dispute, or to unanimously agree on any matter, within its responsibilities, such dispute or matter shall be referred to the JDC for resolution. Either Party may convene a special meeting of the JDC in accordance with Section 2.2(c)(iii) for the purpose of resolving any dispute within the JDC’s jurisdiction that represents a material issue the resolution of which cannot reasonably await until the next scheduled meeting of the JDC.

 

(f)            Dispute Resolution.  If the JDC is unable to resolve any dispute within the responsibilities of the JDC specified in Section 2.2(b), or to unanimously agree on any matter set forth in subsections (i)-(vi) below, within thirty (30) days after a Party provides notice to the other Party of the existence of such dispute, then (A) uniQure shall have final decision-making authority (except as set forth below in this Section 2.2(f)) with respect to all research and Development activities with respect to the Product, with reasonable input from Chiesi taking into account Territory-specific matters, and (B) Chiesi shall have final decision-making authority with respect to all Commercialization activities with respect to the Product in the Territory, with reasonable input from uniQure taking into account uniQure’s global Product strategy; provided, however, that the following decisions must be decided unanimously (or, if not able to be decided unanimously, will be referred to the JSC for resolution pursuant to Section 2.1), in that neither Party shall have the right to exercise its final decision-making authority to unilaterally:

 

(i)        increase the other Party’s obligations or reduce such other Party’s rights under this Agreement, including any obligation to conduct activities, or devote additional personnel to a specific activity to be conducted by such other Party, under the Development Plan and Budget, or require such other Party to conduct activities the costs of which are not reimbursed by such Party or included in Shared Costs;

 

(ii)       expand such Party’s rights or reduce such Party’s obligations under this Agreement;

 

(iii)      resolve disputes regarding the Parties’ rights and obligations under this Agreement;

 

(iv)      make a decision that is expressly stated in this Agreement to require the other Party’s prior approval or consent, or the mutual agreement of the Parties, or that is not consistent with the terms and conditions of this Agreement;

 

(v)       require the other Party to perform any act that such other Party reasonably believes to be inconsistent with Applicable Law; or

 

17



 

(vi)      make a decision in a manner that would cause a breach of any Existing Third Party License or Additional Rights Agreement, or to require any Third Party to take any actions not required to be performed by such Third Party under any Existing Third Party License or Additional Rights Agreement.

 

In addition, each Party shall exercise its final decision-making authority in good faith, subject to the terms and conditions of this Agreement, and in a commercially reasonable manner without favoring other products being developed or commercialized by or on behalf of such Party or its Affiliates outside the Collaboration. With respect to regulatory matters, both Parties agree that they will jointly work towards a regulatory strategy for the Product in the Territory, with an understanding that Chiesi (i) shall have the final decision right on the regulatory strategy for the Product in the Territory, including on all regulatory matters as described in Section 4.1 and Section 4.2 and (ii) shall support uniQure’s global regulatory strategy for the Product unless such support leads to a material increase in costs or time to market for Chiesi, in which case Chiesi shall have the final decision right, as to cause the Parties to maintain the regulatory strategy for the Product in the Territory, as included in the Initial Development Plan and Budget as amended due to any mandatory request of any Regulatory Authority or Applicable Laws in the Territory. For the avoidance of doubt, “material increase in costs” shall mean additional costs (i.e. costs not reimbursed by uniQure to Chiesi) in excess of EUR 2.5 million and “material increase in time to market” shall mean an additional time for submission of a Marketing Authorization Application of more than 180 days, in each case to be determined by comparing such additional costs or additional time with the initial costs or time included in the Initial Development Plan and Budget, as such initial costs or time may have been adjusted due to any mandatory request of any Regulatory Authority or Applicable Laws in the Territory (in other words, any increase of costs or time due to such mandatory requests shall not be relevant for the calculation of the EUR 2.5 million and 180 days thresholds, whereas any other adjustments to the costs or time included in the Initial Development Plan and Budget shall be credited against such thresholds).

 

Without prejudice to the foregoing, it is further understood and agreed that the above final decision-making authority can be exercised by the respective representative(s) of uniQure or Chiesi, as the case may be, also at the JSC level.

 

Section 2.3            Joint Commercialization Committee.  Within thirty (30) days after the Effective Date, Chiesi and uniQure may also establish, as a subcommittee of the JSC, a joint commercialization committee (the “JCC”) to manage any specific matter not otherwise dealt with by the JDC hereunder. If a JCC is formed, the provisions of Sections 2.2(a) and 2.2(c) through (f) above shall apply to the JCC, mutatis mutandis .

 

Section 2.4            Alliance Managers.  Each Party shall appoint an employee (or an employee of its Affiliate) to serve as an alliance manager (“Alliance Manager”) with responsibility for overseeing that the Parties’ activities are conducted in accordance with the Collaboration Agreements, and for being the primary point of contact between the Parties with respect to all such activities. The Alliance Managers to be appointed under this Agreement shall be identical to the ones to be appointed under the Commercialization Agreement. The Alliance Managers are responsible for

 

18



 

driving the Collaboration progress and the resolution of issues between the Parties. The Alliance Managers may be members, but in any event may attend the meetings, of the JSC, JDC, or any other JSC subcommittee, and be responsible for communicating with and reporting to the JSC, JDC, and any other JSC subcommittee, on all relevant matters.

 

ARTICLE III
DEVELOPMENT; MANUFACTURE AND SUPPLY

 

Section 3.1            Overview; Development Plan and Budget.

 

(a)           Subject to and in accordance with the terms and conditions of this Agreement, the Parties shall collaborate on the Development of the Product for the Territory in accordance with the Development Plan and Budget. Each Party shall be responsible for, and shall use Commercially Reasonable Efforts to conduct, those activities assigned to it under the Development Plan and Budget. Unless the Parties otherwise mutually agree:

 

(i)        uniQure shall be responsible for, and shall use Commercially Reasonable Efforts to conduct all activities to Develop the Product in the Territory, including all clinical Development activities that are required to obtain Marketing Authorization in the Territory (with particular emphasis on each Major Country). The Parties will jointly work towards a regulatory strategy for the Product in the Territory, including preparing, filing, obtaining and maintaining all Regulatory Approvals necessary to Develop and Commercialize the Product in each Major Country, subject to Section 2.2(f) and, provided, that all matters under this sub-paragraph (i) shall be included in the Initial Development Plan and Budget; and

 

(ii)       Subject to Section 2.2(f), Chiesi shall be responsible for, and shall use Commercially Reasonable Efforts to conduct all launch and pre-launch activities for the Product in the Territory, including KOL development, market research, and conducting pricing and reimbursement studies.

 

(b)           Each successive Development Plan and Budget shall be prepared by uniQure in consultation with Chiesi, shall be reviewed and approved by the JDC, shall be consistent with the terms and conditions of the Agreement (including this Section 3.1) and shall specify among other things:

 

(i)        Development objectives,

 

(ii)       activities to be performed thereunder for at least the next three (3) years,

 

(iii)      associated budgets for the next year, and good faith projections for the two (2) years thereafter, regarding such activities,

 

(iv)      anticipated timelines for performance, and

 

(v)       specific deliverables.

 

19



 

(c)           The Parties shall update the Development Plan and Budget twice per calendar year and otherwise as reasonably required, as determined by the JDC. uniQure shall propose updates to the Development Plan and Budget in writing to Chiesi at least ten (10) Business Days prior to each of the Regular JDC Meetings. Unless the Parties otherwise agree, the number of FTEs of each Party for a given year of the Development Plan and Budget shall not exceed the applicable number of FTEs set forth in Exhibit F.

 

Section 3.2            Development Reports. Each Party shall provide written reports to the other Party at least ten (10) Business Days before each Regular JDC Meeting, setting forth in reasonable detail such Party’s and its Affiliates’ activities and progress during the six (6) month period ending on the last day of February or August, as applicable, related to the Development of the Product.

 

Section 3.3            Development Program Costs.  The costs of conducting the Development Program shall be borne by the Parties as set forth in Section 8.2.

 

Section 3.4            Manufacture of Product. uniQure shall provide supplies of the Product as necessary for the activities under the Development Plan and Budget; provided, however, that uniQure shall provide commercial supply of the Product pursuant to a supply and distribution agreement (the “HemB Supply and Distribution Agreement”).

 

(a)           Prior to the first dosing of the first patient in a Pivotal Study, the Parties shall negotiate in good faith the HemB Supply and Distribution Agreement, with the terms set forth on Exhibit D and other terms expected to be substantially similar to the Commercialization Agreement; provided, however, that the terms of the HemB Supply and Distribution Agreement, including the “Net Sales” definition and related provisions, may vary from those in the Commercialization Agreement in order to conform to and comply with the Existing Third Party Licenses and any applicable Additional Rights Agreements.

 

(b)           uniQure may supply NIH with inert samples of the Product or its packaging for educational and display purposes.

 

Section 3.5            Failure to Supply.

 

(a)           In the event that it becomes apparent to uniQure that it will be unable to provide supplies of the Product as necessary for the activities under the then current Development Plan and Budget (“Supply Failure”), uniQure shall, immediately after learning of such event or circumstances, notify Chiesi in writing of uniQure’s Supply Failure, along with a reasonable explanation of the reason, to the extent then known to uniQure, for uniQure’s Supply Failure and with a specific indication of the quantity of Product affected by such Supply Failure and anticipated timing of delivery of the Product. Promptly after Chiesi’s receipt of any such notice, the Parties shall agree upon mutually acceptable revised quantities and delivery dates with respect to any ordered Product or, to the extent this is not possible in light of the specific or then unknown reason for uniQure’s Supply Failure, shall discuss in good faith measures to further investigate the root cause and, as the case may be, appropriate steps to overcome such Supply Failure.

 

20



 

(b)                                  Without prejudice to the foregoing paragraph (a), if

 

(i)                                      uniQure’s Supply Failure affects at least supplies for a period of no less than nine (9) months, and

 

(ii)                                   the reason for uniQure’s Supply Failure could be established during the Parties’ discussion pursuant to paragraph (a) above, and such reason was specifically related to uniQure’s ability to Manufacture the Product at its current manufacturing site (i.e. the Supply Failure could reasonably be expected to be overcome if the Product was Manufactured at a different manufacturing site),

 

upon either Party’s request, the Manufacturing of the Product shall be transferred to (A) uniQure’s US manufacturing site, provided such site is operational at the relevant point in time, and further provided uniQure, within one (1) month following such request, does not opt against such transfer, and (B) otherwise (i.e. if uniQure opts against such transfer within the foregoing one (1) month period) to any other Third Party manufacturer mutually agreed to by uniQure and Chiesi. uniQure shall efficiently and promptly transfer to its US manufacturing site or, as the case may be, such Third Party manufacturer all information, licenses and rights controlled by uniQure and necessary to Manufacture the Product during the continuance of uniQure’s Supply Failure. Such transfer shall ensure uniQure’s ongoing control over the information, licenses and right so transferred, shall include the steps outlined in Exhibit G, and shall occur through email and videoconference interactions, as well as face-to-face meetings as required to ensure efficient transfer of technologies and capabilities.

 

If uniQure’s US manufacturing site or, as the case may be, such Third Party manufacturer is unable to Manufacture the Product within fifteen (15) months after uniQure has started the technology transfer to such person, Chiesi shall have the right to terminate this Agreement with three (3) month notice in writing, except if uniQure’s Supply Failure is caused as a result of Force Majeure pursuant to Section 15.7. Such termination shall not become effective if, during such three (3) month notice period, uniQure has notified Chiesi of the ability of its US manufacturing site or, as the case may be, such Third Party manufacturer to Manufacture the Product. Upon termination of this Agreement by Chiesi pursuant to this Section 3.5(b), the provisions of Section 12.6 shall apply.

 

(c)                                   Without prejudice to the foregoing paragraph (a), if

 

(i)                                      uniQure’s Supply Failure affects at least supplies for a period of no less than nine (9) months, and

 

(ii)                                   the reason for uniQure’s Supply Failure (A) could be established during the Parties’ discussion pursuant to paragraph (a) above, but such reason was not specifically related to uniQure’s ability to Manufacture the Product at its current manufacturing site (i.e. the Supply Failure could not reasonably be expected to be overcome if the Product was Manufactured at a different manufacturing site), or (B) could not be established during the Parties’ discussion pursuant to paragraph (a) above during at least the foregoing nine (9) months period, and

 

21



 

(iii)                                uniQure’s Supply Failure is not caused as a result of Force Majeure pursuant to Section 15.7,

 

Chiesi may terminate this Agreement with three (3) month notice in writing. Such termination shall not become effective if, during such three (3) month notice period, uniQure has notified Chiesi of the end of its Supply Failure and has provided to Chiesi at least one of the outstanding orders. Upon termination of this Agreement by Chiesi pursuant to this Section 3.5(c), the provisions of Section 12.6 shall apply.

 

ARTICLE IV
REGULATORY MATTERS

 

Section 4.1                                     Regulatory Filings.  Following the Effective Date, except as may be set forth in the HemB Supply and Distribution Agreement, (a) Chiesi shall own, and shall be responsible for preparing (with input from and in collaboration with uniQure pursuant to Section 3.1 and subject to Section 2.2(f)), filing and maintaining, all regulatory filings and Regulatory Approvals that are required for the Development, use, Manufacture or Commercialization of the Product in the Territory, and (b) Chiesi shall be responsible for all communications and interactions with Regulatory Authorities in the Territory with respect to the Development, use, Manufacture and Commercialization of the Product; provided that, at Chiesi’s reasonable request, uniQure at its expense shall use Commercially Reasonable Efforts to provide assistance to Chiesi in the making of any such regulatory filings.

 

Section 4.2                                     Coordination. Subject to Section 2.2(f), the Parties shall coordinate their regulatory matters with respect to the Product in the Territory taking into account the framework of uniQure’s global regulatory strategy for the Product. In particular:

 

(a)                                  Each Party shall take such actions and otherwise cooperate with the other Party as may be reasonably requested by such other Party to enable such other Party to perform the activities assigned to it as set forth in the Development Plan and Budget, and any other Development or regulatory activities assigned to it under this Agreement, or to comply with any of such other Party’s obligations under the Existing Third Party Licenses or any Additional Rights Agreement.

 

(b)                                  Chiesi shall provide uniQure with electronic copies of all regulatory submissions to, and material communications with, Regulatory Authorities in the Territory and uniQure shall have the right to review and comment on such submissions and communications.

 

(c)                                   Chiesi shall keep uniQure promptly (or to the extent possible, in advance) informed regarding Chiesi’s (or its Affiliate’s) regulatory strategy, planned regulatory submissions and material communications with Regulatory Authorities in the Territory with respect to the Product, including any changes to such strategy, submissions or communications.

 

(d)                                  Chiesi shall provide uniQure with copies of any proposed regulatory submissions to, or material communications with, any Regulatory

 

22



 

Authorities in the Territory with respect to the Product, at least thirty (30) days prior to submission.

 

(e)                                   Chiesi shall promptly provide uniQure with copies of regulatory submissions to, and material communications with, any Regulatory Authorities in the Territory relating to the Product;

 

(f)                                    uniQure shall have the right to have a senior employee of uniQure (expert for each relevant section of CTD) participate in all meetings or substantive teleconferences with the EMA and/or other Regulatory Authorities in the Territory with respect to the Product, as well as to participate in internal meetings or discussions of Chiesi occurring immediately before or after, and related to, such EMA and/or other Regulatory Authorities in the Territory meetings or teleconferences, and shall be provided with advance access to Chiesi’s materials prepared for such EMA and/or other Regulatory Authorities in the Territory meetings and teleconferences.

 

(g)                                   Chiesi shall provide uniQure, if feasible, with reasonable advance notice of any other meeting or substantive teleconference with Regulatory Authorities in the Territory relating to the Product.

 

(h)                                  Without limiting the generality of any of the foregoing in this Section 4.2, Chiesi shall also promptly provide uniQure with an electronic copy of all material correspondence that Chiesi (or its Affiliate) receives from, or submits to, any Regulatory Authority in any country of the Territory related to the Product, including contact reports concerning conversations or substantive meetings, contact reports of all Regulatory Authority interactions in the Territory concerning conversations or substantive meetings, all required periodic reports, and cover letters of all agency submissions (including copies of all attachments to any such cover letters) relating to the Product. Chiesi shall also provide uniQure with any meeting minutes that Chiesi prepares that reflect communications with any Regulatory Authority in any country of the Territory regarding the Product.

 

Section 4.3                                     Product Withdrawals and Recalls. If any Regulatory Authority prior to Marketing Authorization in a country in the Territory (a) threatens, initiates or advises any action to remove the Product from the market in such country, or (b) requires or advises either Party or such Party’s Affiliates to distribute a “Dear Doctor” letter or its equivalent regarding use of the Product in such country, then uniQure or Chiesi, as applicable, shall notify the other Party of such event within three (3) Business Days (or sooner if required by Applicable Law) after such Party becomes aware of the action, threat, advice or requirement. The JDC will meet promptly, but in any case within five (5) Business Days, to discuss and attempt to agree upon whether to recall or withdraw such Product in the Territory; provided, however, that if the Parties fail to agree within an appropriate time period or if the matter involves a safety issue that, in order to protect patient safety, does not allow for sufficient time for a discussion at the JDC level (in which event uniQure shall nonetheless provide advance notice and consultation with Chiesi to the maximum practical extent prior to making a decision), uniQure shall decide whether to recall or withdraw such Product in such country and shall undertake any such recall or withdrawal.

 

Section 4.4                                     Safety Monitoring; Pharmacovigilance.  uniQure shall be responsible for safety monitoring of the Product and for any other obligations

 

23



 

imposed by any Regulatory Authority in connection with the conduct of any preclinical or clinical activities or the granting of the relevant Marketing Authorization. The Parties shall negotiate and execute a detailed safety data exchange agreement (the “SDEA”) prior to the start of clinical Development of the Product, to arrange any future pharmacovigilance exchange between the Parties when relevant . Each Party shall ensure, through its JDC representatives or designated personnel, that the competent pharmacovigilance or clinical groups or personnel from such Party begin to negotiate and establish the appropriate SDEA no later than six (6) months before the start of clinical Development of the Product, or on reasonable request of either Party. The SDEA shall be negotiated in good faith between the pharmacovigilance or clinical groups or personnel of each Party.The SDEA shall define the roles and responsibilities of each Party in terms of pharmacovigilance and define the detailed safety exchange required to permit compliance by each Party with safety reporting requirements to Regulatory Authorities and other entities in the Territory and ensure worldwide safety surveillance. At a minimum, uniQure may share pharmacovigilance information with its licensors and licensees of the Licensed Technology. In the event of a conflict between the terms of this Agreement and the SDEA, the terms of this Agreement shall govern.

 

ARTICLE V
COMMERCIALIZATION

 

Section 5.1                                     Overview.  Subject to the terms and conditions hereunder, including Exhibit D, and the HemB Supply and Distribution Agreement, Chiesi will have sole right and responsibility to Commercialize the Product in the Field in the Territory, including for pre-launch Commercialization activities, including KOL development, pricing and reimbursement studies, as well as for post-launch Commercialization of the Product in the Field in the Territory, including all Chiesi Sole Costs relating thereto, and for booking all sales of the Product throughout the Territory.

 

ARTICLE VI
EXCLUSIVITY

 

Section 6.1                                     Exclusivity.  During the Term, to the fullest extent consistent with any Applicable Law, neither Party nor, subject to Section 15.1, any of such Party’s Affiliates, shall, by itself or through, with or on behalf of any Third Party, undertake the development, manufacture or commercialization anywhere in the Territory of any Competing Product.

 

ARTICLE VII
GRANT OF LICENSES

 

Section 7.1                                     uniQure License Grants.  Subject to the Existing Third Party Licenses and the other terms and conditions of this Agreement, uniQure hereby grants to Chiesi and its Affiliates an exclusive right and license, with the right to grant sublicenses only to Sub-distributors, under the Licensed Technology, to co-Develop, use and Commercialize the Product in the Field in the Territory (the “License”). As used in the preceding sentence, “co-Develop” means that uniQure and its Affiliates may also exercise such Development rights in accordance with this Agreement, but that uniQure shall not grant any such rights to any Third Party in the Territory.

 

24



 

Improvements that are (i) Controlled by uniQure or its Affiliates and (ii) necessary or useful to co-Develop, use or Commercialize the Product in the Field in the Territory, shall be deemed part of the Licensed Technology and included in the License. uniQure shall promptly disclose to Chiesi all Improvements that are Developed by uniQure or its Affiliates (alone or in collaboration with Chiesi or its Affiliates) during the Term and that are necessary or useful to co-Develop, use or Commercialize the Product in the Field in the Territory.

 

Section 7.2                                     Chiesi License Grants.

 

(a)                                  Subject to the terms and conditions of this Agreement, Chiesi hereby grants to uniQure and its Affiliates a non-exclusive, royalty-free, fully paid-up, irrevocable and perpetual (subject to Article XII) license in the Territory, with the right to grant sublicenses, under the Chiesi Technology, for the purposes of conducting Development Program activities.

 

(b)                                  Subject to the terms and conditions of this Agreement, Chiesi hereby grants to uniQure a non-exclusive, worldwide, royalty-free, fully paid-up, irrevocable, perpetual (subject to Article XII) license, with the right to grant sublicenses, under the Chiesi Technology (including, subject to Section 7.3 any rights Chiesi may have in and to the Product Data), to Develop, Manufacture, having Manufactured, use, and Commercialize (outside the Territory) the Product in the Field.

 

Section 7.3                                     Use of Product Data Outside of Territory.  Chiesi shall provide uniQure, promptly following uniQure’s request, with any Product Data not otherwise in uniQure’s possession, and uniQure shall have the right to use such Product Data in connection with development and regulatory activities conducted by or on behalf of uniQure outside the Territory.

 

Section 7.4                                     Disclosure of and License under Chiesi Improvements.  Chiesi shall promptly disclose to uniQure all Improvements that are developed by Chiesi or its Affiliates (alone or in collaboration with uniQure or its Affiliates) during the Term. Chiesi shall, and hereby does, grant to uniQure a non-exclusive, worldwide, royalty-free, fully paid-up, irrevocable and perpetual (subject to Article XII) license, with the right to sublicense, under all Improvements Controlled by Chiesi and its Affiliates, to Develop, Manufacture, having Manufactured, use, and Commercialize (outside the Territory) the Product in the Field.

 

Section 7.5                                     Compliance with Third Party Agreements.

 

(a)                                  The grants by uniQure under Licensed Technology set forth in Section 7.1 include the sublicense of certain Licensed Technology that is not owned by uniQure. Chiesi’s rights and licenses under, or with respect to, Licensed Technology, including any Patent Prosecution or enforcement undertaken by the Parties pursuant to Article IX, are limited to the rights granted by Third Party licensors to uniQure under the Existing Third Party Licenses and are subject to all applicable restrictions, limitations and obligations imposed on uniQure or its sub-licensees in such Existing Third Party Licenses. Chiesi shall comply, and cause its Affiliates and Sub-distributors to comply, with all such restrictions, limitations and obligations mutatis mutandis (including Paragraphs 5.1-5.4, 8.1, 10.1, 10.2, 12.5, and

 

25



 

13.8-13.10 of the PHS 2011 Agreement, and Paragraphs 5.1, 5.2, 8.1, 10.1, 10.2, 12.5 and 13.6-13.8 of the PHS 2007 Agreement, a copy of which provisions is attached hereto as Exhibit B). To the extent there is a conflict between the terms of any Existing Third Party License and the rights granted to Chiesi hereunder, the terms of such Existing Third Party License shall control solely with respect to the Patent Rights and Know-How owned or controlled by the applicable Third Party licensor. Notwithstanding anything to the contrary in this Agreement, either Party may not exercise any of its rights under this Agreement (including any right to any cure period (including under Section 12.3) or to delay performance of an obligation (including under Article XIII or Section 15.7)) in any manner that would result in any licensor having a right to terminate an Existing Third Party License, or that would cause the other Party to be in breach of any of its obligations under any Existing Third Party License.

 

(b)                                  During the Term, uniQure shall comply with the Existing Third Party Licenses in effect which are then applicable to the activities under this Agreement with respect to the Product (and in particular shall not commit any breach that would entitle the Third Party licensor to terminate such an Existing Third Party License) and shall not terminate any such Existing Third Party License without Chiesi’s prior written consent. In addition, during the Term, uniQure shall promptly notify Chiesi of any written notice of breach or termination received by uniQure with respect to any such Existing Third Party License and, to the extent that uniQure does not cure such breach at least ten (10) Business Days before the date on which the relevant licensor could terminate such Existing Third Party License due to such breach by uniQure, Chiesi shall have the right (to the extent consistent with such Existing Third Party License) to cure any such breach on uniQure’s behalf and in such a case, Chiesi shall have the right to deduct (i) any and all arm’s length payments made on behalf of uniQure for the above purpose, from the next due payments to be made hereunder plus (ii) interest on such payments calculated pursuant to Section 8.5 below.

 

(c)                                   Any sublicensee obligations required by any Existing Third Party License to be included in a sublicense thereunder, including any required provision making the applicable Third Party licensor a third party beneficiary of any sublicense thereunder, shall be deemed to be included in this Agreement; provided that, a copy of the relevant agreement provisions has been attached hereto as Exhibit B.

 

(d)                                  The license granted by uniQure in Section 7.1 with respect to the Patent Rights licensed under the Existing Third Party Licenses are subject to rights reserved by the licensors and the US government as set forth in the Existing Third Party Licenses.

 

Section 7.6                                     Additional Rights Acquired after Effective Date.

 

(a)                                  During the Term, if either Party identifies the need for, or is otherwise offered, a license, covenant not to sue or similar rights to any Third Party Patent Right or Third Party Know-How that such Party in good faith believes is necessary or useful for the Development, use, Manufacture, having Manufactured, or Commercialization of the Product in the Field in the Territory (“Additional Rights”), then such Party shall promptly notify the other Party and, in any event, prior to

 

26



 

commencing negotiation or entering into an agreement with respect to such Additional Rights, and the Parties’ rights to conduct such negotiations shall be subject to the remaining provisions of this Section 7.6. The Parties shall thereafter conduct good faith discussions regarding whether such Additional Rights are necessary or useful for the Development, use, Manufacture, having Manufactured, or Commercialization of the Product in the Field in the Territory or whether they otherwise agree that such Additional Rights should be acquired.

 

(b)                                  uniQure shall have the first right (but not the obligation) to license or otherwise acquire rights to any Additional Rights. If uniQure provides written notice to Chiesi that uniQure declines to exercise such first right, then Chiesi shall have the right (but not the obligation) to pursue acquiring rights to any given Additional Rights. The Party pursuing any given Additional Rights (the “Controlling Party”) shall keep the other Party (the “Non-Controlling Party”) reasonably informed regarding the status thereof and shall use Commercially Reasonable Efforts to obtain from the applicable Third Party licensor the right to sublicense such Additional Rights under the licenses granted to the Non-Controlling Party hereunder.

 

(c)                                   If the Controlling Party acquires rights to any Additional Rights and has the right to grant a sublicense under such Additional Rights to the Non-Controlling Party, and the Non-Controlling Party wishes to include such Additional Rights in the licenses granted to the Non-Controlling Party hereunder (or under the HemB Supply and Distribution Agreement), the Non-Controlling Party shall notify the Controlling Party of its desire to do so and the Controlling Party shall provide the Non-Controlling Party a summary of all material restrictions on the scope of the licenses granted under, and all material payment obligations that would be owed by the Non-Controlling Party with respect to, any Third Party agreement applicable to such Additional Rights. The Non-Controlling Party may, upon written notice to the Controlling Party and subject to Section 7.6(d), Section 7.6(e) and Section 7.6(f), obtain a sublicense under such Additional Rights and include such Additional Rights under the licenses granted to the Non-Controlling Party hereunder.

 

(d)                                  Following such notice from the Non-Controlling Party that it desires to include any given Additional Rights under the license granted to the Non-Controlling Party hereunder (or under the HemB Supply and Distribution Agreement), (i) any such Additional Rights that do not carry financial or other obligations or restrictions shall be included automatically under the applicable license hereunder, and (ii) subject to Section 7.6(e) below, any such Additional Rights that carry financial or other obligations or restrictions shall be included under the applicable license hereunder only if the Non-Controlling Party agrees to share the costs of such Additional Rights (including any upfront payment or similar acquisition cost to access such Additional Rights) with the Controlling Party and to assume all other obligations to, and be subject to all restrictions imposed by, the Controlling Party’s licensor to the extent arising from the grant to the Non-Controlling Party under such Additional Rights (including, to the extent access to such terms have been made available to the Non-Controlling Party in unredacted form, all other terms of the Additional Rights Agreement that apply to the licenses granted to the Non-Controlling Party hereunder).

 

27



 

(e)                                   If the Parties are unable, after twenty (20) Business Days, to agree as to whether any given Additional Rights are in fact necessary or useful for the Development, use, Manufacture, having Manufactured, or Commercialization of the Product in the Field in the Territory or if the Parties are unable to agree to the allocation of the costs (as specified above), then, notwithstanding Article XIII, the Parties shall jointly engage an expert panel consisting of patent attorney(s) or expert(s) in the development, manufacturing or commercialization of products comparable to the Product in question and other CMC matters, as applicable, not regularly employed by either Party to resolve such dispute. The decision of such expert panel shall be binding on the Parties as to such dispute.

 

(f)                                    Nothing in this Section 7.6 shall restrict either Party, at such Party’s sole cost and expense, from licensing or otherwise acquiring any additional rights that are not necessary or useful for the Development, use, Manufacture, having Manufactured, or Commercialization of the Product in the Field in the Territory.

 

Section 7.7                                     No Implied Licenses.  Except as explicitly set forth in this Agreement, neither Party grants to the other Party any license, express or implied, under its intellectual property rights.

 

ARTICLE VIII
FINANCIAL PROVISIONS

 

Section 8.1                                     Upfront Payment.  Subject to the condition precedent pursuant to Section 12.1(b),

 

(a)                                  in recognition of uniQure’s past expenditure developing the Product, Chiesi shall pay uniQure a one-time, non-refundable, non-creditable fee of Five Million Euros (EUR 5,000,000);

 

(b)                                  as reimbursement of uniQure’s past expenditure setting up the Manufacturing site for the Product in The Netherlands, Chiesi shall pay uniQure a one-time, non-refundable, non-creditable amount of Seven Million Five Hundred Thousand Euros (EUR 7,500,000); and

 

(c)                                   in consideration of uniQure making available to Chiesi Manufacturing capacity at uniQure’s Manufacturing site for the Product under this Agreement, Chiesi shall pay uniQure a one-time, non-refundable, non-creditable fee of Two Million Five Hundred Thousand Euros (EUR 2,500,000);

 

(a) to (c), collectively, the “Upfront Payment”.

 

Chiesi shall pay the Upfront Payment to uniQure within ten (10) Business Days after this Agreement has become effective pursuant to Section 12.1(b), provided receipt of a proper invoice from uniQure for the Upfront Payment.

 

Section 8.2                                     Development Program Costs.

 

(a)                                  Shared Costs.

 

28



 

(i)                                      At the end of each calendar quarter, in accordance with paragraph (d) below, Chiesi shall pay to uniQure fifty percent (50%) of the actual Shared Costs incurred by uniQure during such calendar quarter (including, after the end of the first calendar quarter, the actual Shared Costs incurred by uniQure during the period prior to the date this Agreement has become effective pursuant to Section 12.1(b)), and uniQure shall pay to Chiesi fifty percent (50%) of the actual Shared Costs incurred by Chiesi during such calendar quarter. To this end, each Party shall provide to the other Party and to the JDC, within twenty (20) days after the end of each calendar quarter, (A) a written report with an accounting of and copies of supporting invoices for Shared Costs actually incurred by such Party during such calendar quarter (including, after the end of the first calendar quarter, the actual Shared Costs incurred by uniQure during the period prior to the date this Agreement has become effective pursuant to Section 12.1(b)) or, with respect to Third Party invoices not timely received by a Party, for Shared Costs incurred with respect to previous calendar quarters (such amount, “uniQure Shared Cost” or “Chiesi Shared Cost”, as applicable), (B) an estimate of the Shared Costs to complete the current year of the Development Program (which shall be the estimated cost to complete the current year of the Initial or newly agreed Development Plan and Budget), and (C) an estimate of the Shared Costs to complete the Development Program (which shall be the estimated cost to complete the Initial or newly agreed Development Plan and Budget).

 

(ii)                                   In the case the estimated Shared Costs to complete the Development Program as set out in sub-paragraph (a)(i)(C) above is equal or within a ten percent (10%) budget overrun of the Initial or newly agreed Development Plan and Budget, Chiesi or, as the case may be, uniQure agrees to make the quarterly payment pursuant to paragraph (i) in full. In case the estimated Shared Costs to complete the Development Program as set out in sub-paragraph (a)(i)(C) above is more than ten percent (10%) higher than the Initial or newly agreed Development Plan and Budget, Chiesi and uniQure shall investigate the cause of this budget overrun and shall jointly agree to next steps (in the JDC). Any activity that is not budgeted in the Initial or newly agreed Development Plan and Budget shall be handled as a scope change and each scope change (incl. the budget of the scope change) must be agreed beforehand by both Parties and, when agreed, the Development Plan and Budget shall be deemed amended accordingly. Without prejudice to the foregoing provisions of this sub-paragraph (ii), any above budget overrun of the Initial or newly agreed Development Plan and Budget or any new activity not budgeted but agreed beforehand by both Parties, shall be calculated at the actual direct costs therefor (including costs for out-of-pocket expenses as well as costs for personnel at actual direct costs), without any reference to the FTE Rate.

 

(iii)                                Within ten (10) Business Days after this Agreement has become effective pursuant to Section 12.1(b), Chiesi agrees to pay EUR 250,000 as a first payment to prevent uniQure of pre-paying the activities of the Initial Development Plan and Budget. At the end of the Development Program, uniQure shall credit the last quarterly payment against such EUR 250,000 payment and pay any remaining amount, if any, to Chiesi within sixty (60) days after the end of such last calendar quarter.

 

29



 

(b)                                  Chiesi Sole Costs.  uniQure shall provide to Chiesi and to the JDC, within twenty (20) days after the end of each calendar quarter, a written report with an accounting of and copies of supporting invoices for Chiesi Sole Costs actually incurred by uniQure during such calendar quarter or, with respect to Third Party invoices not timely received by uniQure, for Chiesi Sole Costs incurred with respect to previous calendar quarters.

 

(c)                                   uniQure Sole Costs.  Chiesi shall provide to uniQure and to the JDC, within twenty (20) days after the end of each calendar quarter, a written report with an accounting of and copies of supporting invoices for uniQure Sole Costs actually incurred by Chiesi during such calendar quarter or, with respect to Third Party invoices not timely received by Chiesi, for uniQure Sole Costs incurred with respect to previous calendar quarters.

 

(d)                                  Reconciliation Payment. Within twenty (20) days after receipt of Chiesi’s accountings of Chiesi Shared Cost pursuant to Section 8.2(a)(i) and of uniQure Sole Costs pursuant to Section 8.2(c), uniQure shall calculate the amount of a payment (the “Reconciliation Payment”) necessary to effect the Parties’ agreed allocation of costs as follows:

 

(i)                          each Party shall bear fifty percent (50%) of all Shared Costs;

 

(ii)                       Chiesi shall bear one hundred percent (100%) of all Chiesi Sole Costs; and

 

(iii)                    uniQure shall bear one hundred percent (100%) of all uniQure Sole Costs;

 

pursuant to the following calculation:

 

Reconciliation Payment =

 

(.5 × uniQure Shared Costs)
– (.5 × Chiesi Shared Costs)
+ Chiesi Sole Costs reported by uniQure pursuant to Section 8.2(b)
– uniQure Sole Costs reported by Chiesi pursuant to Section 8.2(c).

 

Promptly following such calculation, uniQure shall provide to Chiesi a written report thereof. Such report shall be accompanied, as applicable, by either an invoice (if the Reconciliation Payment is positive) or a credit memo (if the Reconciliation Payment is negative), which credit memo Chiesi may apply to any future payment due to uniQure under this Section 8.2. Chiesi shall pay any such invoice within sixty (60) days after receipt.

 

Section 8.3                                     Recordkeeping; Audit Rights.  Each Party shall keep, and shall require its Affiliates to keep, complete and accurate records of the latest eight (8) years of costs incurred by such Party in the conduct of Development and regulatory activities under the Development Plan and Budget (including the activities set forth in

 

30



 

the Initial Development Plan and Budget). For the sole purpose of verifying costs included in the reports provided pursuant to Section 8.2, each Party shall have the right annually (but no more than once per year) at such Party’s expense to retain an independent certified public accountant selected by such Party, and reasonably acceptable to the other Party, to review such records in the location(s) where such records are maintained by the other Party or its Affiliates upon reasonable notice and during regular business hours and under obligations of confidence. Results of such review shall be made available to both Parties. If the review indicates that there was an underpayment of any amount payable to the auditing Party, the amount of such underpayment shall be remitted to the auditing Party within sixty (60) days after such review, together with interest calculated in the manner provided in Section 8.5.If the underpayment is equal to or greater than five percent (5%) of the amount that was otherwise due, the audited Party shall pay all of the auditing Party’s reasonable out-of-pocket expenses of such review. If the review indicates that there was an overpayment of any amounts by the audited Party, the audited Party may apply the amount of such overpayment to any future payment due to the auditing Party under Section 8.2.

 

Section 8.4                                     Method of Payment.  All amounts payable by a Party (the “Paying Party”) hereunder shall be paid by or on behalf of such Paying Party in Euros. Shared Costs, uniQure Sole Costs or Chiesi Sole Costs incurred in a currency other than Euros shall be expressed in their Euro equivalent, calculated on the last Business Day of the calendar quarter to which the applicable report relates using the currency converter at www.oanda.com. All payments due to a Party (the “Receiving Party”) hereunder shall be made by wire transfer directly to an account designated by the Receiving Party in writing. The Receiving Party shall be responsible for all charges from the receiving bank due to the receipt of the wire transfer. The Paying Party shall be responsible for all other bank costs.

 

Section 8.5                                     Late Payments.  Any payment under this Agreement that is not paid on or before the date such payment is due shall bear interest at the lesser of (a) three (3) months of EURIBOR rate plus three percent (3%) per year, or (b) the highest rate permitted by Applicable Laws, calculated on the number of days such payments are overdue.

 

Section 8.6                                     Tax Withholding.  To the extent that any payments hereunder by the Paying Party to the Receiving Party are subject to tax, the Paying Party shall pay such tax; provided, however, that, with respect to any payments subject to withholding tax, the Paying Party shall pay the applicable withholding tax amount to the relevant taxing authority and promptly provide the Receiving Party with all necessary documentation for the Receiving Party to recover such tax. The Paying Party will take all reasonable and lawful steps to minimize the amount of any such withholding tax obligation and the Receiving Party shall promptly provide all information and documentation in its possession necessary for doing so.

 

Section 8.7                                     Blocked Payments.  In the event that, by reason of Applicable Laws in any country, it becomes impossible or illegal for the Paying Party to transfer payments to the Receiving Party, the Paying Party shall, to the extent consistent with Applicable Laws, have such royalties or other payments paid to the Receiving Party by an Affiliate of the Paying Party. To the extent such payment is not consistent with

 

31



 

Applicable Laws, the Paying Party shall deposit such payments in local currency in the relevant country to the credit of the Receiving Party in a recognized banking institution designated by the Receiving Party or, if none is designated by the Receiving Party within a period of thirty (30) days after written request from the Paying Party, in a recognized banking institution selected by the Paying Party and identified in a notice in writing given to the Receiving Party.

 

ARTICLE IX
INTELLECTUAL PROPERTY

 

Section 9.1                                     Ownership.

 

(a)                                  Existing Intellectual Property.  Except as expressly set forth in this Agreement and subject to the licenses granted under this Agreement, as between the Parties each Party shall retain all right, title and interest in and to the Patent Rights, Know-How and other intellectual property rights owned by or Controlled by such Party or its Affiliates as of the Effective Date.

 

(b)                                  Solely Owned Know-How.  Except as expressly set forth in this Agreement and subject to the licenses granted by such Party under this Agreement, as between the Parties each Party or its Affiliate, as applicable, shall exclusively own all right, title and interest in and to all Know-How made or conceived solely by the employees, agents or consultants of such Party or its Affiliates in the course of performing its activities under this Agreement.

 

(c)                                   Joint Know-How.  All Know-How made or conceived jointly by employees, agents or consultants of uniQure or its Affiliates, on the one hand, and employees, agents or consultants of Chiesi or its Affiliates, on the other hand, shall be owned jointly on the basis of each Party having an undivided one-half (½) interest in the whole (“Joint Know-How”), and each Party hereby assigns to the other Party a sufficient interest in its rights in and to the Joint Know-How so as to effect such joint ownership. Subject to the licenses granted herein and each Party’s payment obligations hereunder, each Party shall have the right to exploit the Joint Know-How, or sell, license or otherwise transfer or grant rights under Joint Know-How, or any Joint Patents directed to the Joint Know-How, to its Affiliates or any Third Party, without any duty to account to the other Party; provided that, during the Term neither Party nor its Affiliates may use, sell, license or otherwise transfer or grant rights under Joint Know-How, or any Joint Patents directed to such Joint Know-How, to any Affiliate or Third Party in any manner which would conflict with, or limit the scope of, any of the rights or licenses granted to the other Party hereunder.

 

(d)                                  Inventorship.  For purposes of determining the Parties’ rights under this Agreement, the determination of inventorship shall be made in accordance with US patent laws.

 

Section 9.2                                     Prosecution and Maintenance of Patent Rights.

 

(a)                                  Licensed Patents.  Subject to any rights of and obligations to uniQure’s Third Party licensors, uniQure shall have the exclusive right and obligation to conduct Patent Prosecution for the Licensed Patents (other than any Joint Patent) in the Territory, in uniQure’s name, and to control any interference, derivation

 

32



 

proceeding, reexamination, review, opposition and similar proceedings relating thereto. Subject to any rights of and obligations to uniQure’s Third Party licensors, uniQure shall promptly and regularly inform and consult with Chiesi regarding the Patent Prosecution, including any interference, derivation proceeding, reexamination, review, opposition and similar proceedings relating thereto, of all Licensed Patents in the Territory.

 

(b)                                  Chiesi Patents.  Chiesi shall have the exclusive right and obligation to conduct Patent Prosecution for the Chiesi Patents (other than any Joint Patents) in Chiesi’s name, and to control any interference, derivation proceeding, reexamination, review, opposition and similar proceedings relating thereto. Chiesi shall promptly and regularly inform and consult with uniQure regarding the Patent Prosecution, including any interference, derivation proceeding, reexamination, review, opposition and similar proceedings relating thereto, of all Chiesi Patents.

 

(c)                                   Joint Patents.  uniQure shall have the first right and option (but not the obligation) to conduct Patent Prosecution for the Joint Patents in uniQure’s name, and to control any interference, derivation proceeding, reexamination, review, opposition and similar proceedings relating thereto. In the event that uniQure elects to conduct Patent Prosecution according to the foregoing sentence, uniQure shall grant, and hereby grants, to Chiesi, subject to the terms and conditions of this Agreement, a non-exclusive, worldwide, royalty-free, fully paid-up, irrevocable, perpetual license, with the right to grant sublicenses, under the Joint Patents to Develop, use, Manufacture, have Manufactured, and Commercialize the Product in the Field. In the event that uniQure elects not to conduct Patent Prosecution for, or elects to abandon, any Joint Patent, or declines to control any related interference, derivation proceeding, reexamination, review, opposition or similar proceedings, uniQure shall give Chiesi reasonable written notice to this effect, sufficiently in advance to permit Chiesi, in its sole discretion, to undertake such Patent Prosecution, or to control such interference, derivation proceeding, reexamination, review, opposition or similar proceedings, without a loss of rights, and thereafter Chiesi may, upon written notice to uniQure and in Chiesi’s name, conduct Patent Prosecution for such Joint Patents and control such interference, derivation proceeding, reexamination, review, opposition or similar proceedings. If required under Applicable Laws in order for the prosecuting Party to control any interference, derivation proceeding, reexamination, review, opposition and similar proceedings relating to any Joint Patent, the other Party shall join as a party to such interference, derivation proceeding, reexamination, review, opposition and similar proceedings.

 

(d)                                  Cooperation.  Each Party agrees to cooperate with the other Party with respect to Patent Prosecution, including any interference, derivation proceeding, reexamination, review, opposition and similar proceedings relating thereto, of Joint Patents pursuant to Section 9.2(c), subject to any rights of, and obligations to, uniQure’s Third Party licensors, including by:

 

(i)                          executing all such documents and instruments and performing of such acts as may be reasonably necessary in order to permit the other Party to continue any Patent Prosecution that such Party has elected not to pursue, as provided for in Section 9.2(c);

 

33



 

(ii)                       making its employees, agents and consultants reasonably available to the other Party (or to the other Party’s authorized attorneys, agents or representatives), to the extent reasonably necessary to enable the prosecuting Party to undertake Patent Prosecution;

 

(iii)                    providing (itself or through patent counsel) the other Party with a copy of each proposed material correspondence pertaining to substantive Patent Prosecution on the merits with World Intellectual Property Office (“WIPO”) or the European Patent Office (“EPO”), as well as providing draft copies of patent applications to be submitted to the WIPO under the Patent Cooperation Treaty, or submitted to any patent office in the Territory in a form substantially different from that previously submitted to the WIPO, reasonably in advance of any applicable filing or response deadline to allow the other Party to review and comment on the content of such proposed correspondence and advise the prosecuting Party as to the conduct of such Patent Prosecution, which comments and advice the prosecuting Party will consider in good faith;

 

(iv)                   providing (itself or through patent counsel) the other Party with copies of all material correspondence pertaining to substantive Patent Prosecution on the merits with the WIPO or the EPO after its submission or receipt, as the case may be; and

 

(v)                      seeking patent term extensions, adjustments, and the like wherever available for the Product.

 

Section 9.3                                     Third Party Infringement.

 

(a)                                  Notice.  Each Party shall promptly report in writing to the other Party during the Term any known or suspected infringement of any Valid Claims within the Licensed Patents, Chiesi Patents or Joint Patents involving the use, manufacture or commercialization of a product or product candidate that is or would likely be a Competing Product (“Competitive Infringement”), and shall provide the other Party with all available evidence supporting such infringement or suspected infringement. Promptly after receipt of a notice of a Competitive Infringement in the Territory, the Parties shall discuss in good faith the infringement and appropriate actions that could be taken to cause such Competitive Infringement to cease.

 

(b)                                  Enforcement of Patents.  The Party with responsibility under Section 9.2 for Patent Prosecution of the Patent Right that is subject of the Competitive Infringement shall have the exclusive right to initiate a suit or take other appropriate action that it believes is reasonably required to prevent or abate actual or threatened infringement of, or otherwise enforce, in the best commercial interests of the Product, the applicable Patent Right. The Party filing any such suit or taking any such action shall control all decision making related to any such suit or action, subject to Section 9.3(c) below.

 

(c)                                   Conduct of Actions.  The Party initiating suit or action pursuant to Section 9.3(b) with respect to Competitive Infringement in the Territory shall have the sole and exclusive right to select counsel for such suit or action. At the initiating Party’s request and expense, the other Party shall join as a party to the suit or action. Such other Party shall offer reasonable assistance to the initiating Party in connection

 

34



 

therewith. The initiating Party shall provide the other Party with an opportunity to make suggestions and comments regarding such suit or action. The initiating Party shall, to the extent permitted by Applicable Laws, keep the other Party promptly informed, and shall from time to time consult with such other Party, regarding the status of any such suit or action and shall provide such other Party with copies of all material documents (including complaints, answers, counterclaims, material motions, orders of the court, memoranda of law and legal briefs, interrogatory responses, depositions, material pre-trial filings, expert reports, affidavits filed in court, transcripts of hearings and trial testimony, trial exhibits and notices of appeal) filed in, or otherwise directly relating to, such suit or action. The Party not initiating such suit or action shall have the right to participate and be represented in any such suit by its own counsel at its own expense. Neither Party shall conduct any such suit or action in a manner that materially places at risk the scope or validity of any Joint Patent, and neither Party shall settle or compromise any claim or proceeding relating to any Joint Patent, without obtaining the prior written consent of the other Party. If Chiesi or any of its Affiliates conduct any such suit or action in a manner that materially places at risk the scope or validity of any Licensed Patent, uniQure may terminate this Agreement in accordance with the provisions of Section 12.4. If uniQure or any of its Affiliates conduct any such suit or action in a manner that materially places at risk the scope or validity of any Chiesi Patent, Chiesi may terminate this Agreement in accordance with the provisions of Section 12.4.

 

(d)                                  Recoveries.  With respect to any suit or action to protect any Joint Patent referred to in Section 9.3(b) above, any recovery obtained by a Party as a result of any such proceeding, by settlement or otherwise, shall be applied in the following order of priority:

 

(i)                          first, such Party shall pay to the applicable licensor any amount to which such licensor is entitled pursuant to the terms of any Existing Third Party License or Additional Rights Agreement; and

 

(ii)                       second, any remainder shall be allocated equally between the Parties.

 

Section 9.4                                     Claimed Infringement.  In the event that a Party becomes aware of any claim or threat of claim that the Development, use, Manufacture, have Manufactured or Commercialization hereunder of the Product infringes or misappropriates the intellectual property rights of any Third Party, such Party shall promptly notify the other Party. Each Party shall provide to the other Party copies of any notices such Party receives from Third Parties regarding any alleged infringement of Third Party Patent Rights or any alleged misappropriation of Third Party Know-How. Such notices shall be provided promptly, but in no event after more than fifteen (15) days following receipt thereof. In any such instance, the Parties shall cooperate in undertaking an appropriate course of action.

 

Section 9.5                                     Patent Invalidity Claim. If a Third Party at any time asserts a claim that any Licensed Patent (including a Joint Patent) or Chiesi Patent (including a Joint Patent) that covers the composition of matter of the Product or the method of use of the Product in the Field in the Territory is invalid or otherwise unenforceable (“Invalidity Claim”), either as a defense in an infringement action brought by Chiesi or uniQure pursuant to Section 9.3 or in an action brought against Chiesi or uniQure

 

35



 

under Section 9.4, including any declaratory judgment action, the Parties shall cooperate with each other in (i) preparing and formulating a response to such Invalidity Claim and (ii) undertaking any further and appropriate course of action. Neither Party shall settle or compromise any Invalidity Claim without the consent of the other Party, which consent shall not be unreasonably withheld.

 

Section 9.6                                     Licensor Rights.  All obligations under Sections 9.2 through 9.5 are subject to the rights of the relevant licensor pursuant to the terms of any Existing Third Party License or Additional Rights Agreement.

 

ARTICLE X
CONFIDENTIALITY

 

Section 10.1                              Confidential Information.  All Confidential Information disclosed by a Party or any of its Affiliates to the other Party or any of its Affiliates before or during the Term shall not be used by the receiving Party or any of its Affiliates except in connection with the activities contemplated by this Agreement, shall be maintained in confidence by the receiving Party and its Affiliates, and shall not otherwise be disclosed by the receiving Party or its Affiliates to any Third Party (except as set forth in the remainder of this Article X), without the prior written consent of the disclosing Party, except to the extent that the Confidential Information:

 

(a)                                  was known or used by the receiving Party or any of its Affiliates prior to its date of disclosure by the disclosing Party;

 

(b)                                  either before or after the date of the disclosure to the receiving Party hereunder or under the Confidentiality Agreement is lawfully disclosed to the receiving Party or any of its Affiliates by a Third Party rightfully in possession of and with the right to disclose such Confidential Information other than under an obligation of confidentiality;

 

(c)                                   either before or after the date of the disclosure to the receiving Party hereunder or under the Confidentiality Agreement becomes generally known to the public through no fault or omission on the part of the receiving Party or its Affiliates;

 

(d)                                  is independently developed by or for the receiving Party or any of its Affiliates without reference to or reliance upon any of the other Party’s Confidential Information; or

 

(e)                                   is required to be disclosed by the receiving Party or its Affiliates to comply with Applicable Laws, which may include the rules of Euronext, of the US Securities and Exchange Commission (“SEC”), or of any other stock exchange, or to defend or prosecute litigation or arbitration or to comply with legal process; provided that, the receiving Party provides prior written notice of such disclosure to the disclosing Party (to the extent feasible) and only discloses Confidential Information of the other Party to the extent necessary for such legal compliance or litigation purpose; and provided, further, that such information shall otherwise remain Confidential Information (subject to the exceptions in this Section 10.1).

 

36



 

Notwithstanding the foregoing, clauses (a), (b) and (d) shall not alter the requirement to keep the terms and conditions of this Agreement confidential, as set forth herein, subject to the remainder of this Article X.

 

Section 10.2                              Employee, Director, Consultant and Advisor Obligations.  Chiesi and uniQure each agrees that it and its Affiliates shall provide Confidential Information received from the other Party only to the receiving Party’s employees, directors, consultants, agents and advisors, and to the employees, directors, consultants, agents and advisors of the receiving Party’s Affiliates, who have a need to know such Confidential Information to assist the receiving Party in fulfilling its obligations under this Agreement and who are bound by obligations of confidentiality and non-use that are at least as restrictive as those set forth in this Agreement. Each Party shall remain responsible for any failure by any of its or its Affiliates’ employees, directors, consultants, agents and advisors to treat such Confidential Information as required under this Article X.

 

Section 10.3                              Publicity.

 

(a)                                  Following execution of this Agreement, the Parties shall jointly or separately issue a press release, in a text to be agreed upon between the Parties in advance, announcing the execution of this Agreement and the Commercialization Agreement.

 

(b)                                  Thereafter, each Party shall only issue press releases (other than the press release pursuant to paragraph (a) above) or make other public disclosures regarding this Agreement or the Parties’ activities under this Agreement (each such press release or public disclosure, a “Subject Disclosure”):

 

(i)                                      that have been approved in writing in advance by the other Party (such approval not to be unreasonably withheld, conditioned or delayed);

 

(ii)                                   if advised by counsel to issue such Subject Disclosure in order to comply with Applicable Laws, which may include the disclosure rules of SEC or a similar regulatory agency in a country in the Territory or of Euronext or any other stock exchange of other securities trading institution (for clarity such issuance is also subject to Section 10.3(c));

 

(iii)                                subject to Section 10.3(c), if the contents of such Subject Disclosure have previously been made public other than through a breach of this Article X by a Party; or

 

(iv)                               subject to sub-paragraph (i) above, to the extent that such Subject Disclosure describes one or more of the following:

 

(A)                    preclinical results with respect to the Product;

 

(B)                    the commencement, completion or “top-line” results of clinical studies of the Product;

 

(C)                    the completion of patient enrollments for clinical studies of the Product;

 

37



 

(D)                    the filing for or receipt of Marketing Authorization with respect to the Product;

 

(E)                     the Patent Prosecution or enforcement of any of the Licensed Patents, including the issuance of any patent included in the Licensed Patents;

 

(F)                      the receipt of any regulatory exclusivity for the Product; or

 

(G)                    the first Party’s presence or participation at scientific, financial or investor forums.

 

(c)                                   Unless not feasible under the circumstances because of the need to comply with Applicable Laws or stock exchange rules, the Party making a Subject Disclosure shall provide the other Party with a draft Subject Disclosure at least ten (10) Business Days prior to its intended publication for the other Party’s review. Such other Party may provide the first Party with suggested modifications to the draft Subject Disclosure. The first Party shall consider in good faith the other Party’s suggestions in issuing such Subject Disclosure.

 

(d)                                  For clarity, nothing in this Agreement shall restrict each Party from issuing press releases or making other public disclosures regarding such Party’s development, manufacturing or commercialization activities with respect to any product other than the Product, or, with reference to uniQure only, with respect to any Product outside the Territory.

 

Section 10.4                              Other Disclosures.  Notwithstanding anything in this Agreement to the contrary, each Party shall have the right to disclose the other Party’s Confidential Information (including the terms of this Agreement) (as applicable):

 

(a)                                  to such Party’s then-current or potential investors, lenders, acquirers, investment bankers, and other Third Parties in connection with financing, partnering (to the extent consistent with this Agreement) and acquisition activities, solely on a need-to-know basis and under obligations of confidentiality and non-use that are at least as restrictive as those set forth in this Article X;

 

(b)                                  as required by the Existing Third Party Licenses or any Additional Rights Agreement;

 

(c)                                   to conduct Patent Prosecution or enforcement of Patent Rights for which such Party is responsible hereunder;

 

(d)                                  to such Party’s then-current or potential collaborators, and Third Party contractors (including contract manufacturers and Sub-distributors) for purposes of engaging in the Development, use, Manufacture or Commercialization of the Product as contemplated hereunder, solely on a need-to-know basis and under obligations of confidentiality and non-use that are at least as restrictive as those set forth in this Article X.

 

38



 

Section 10.5                              Publications.

 

(a)                                  Notwithstanding Section 10.3 and Section 10.4, a Party (the “Publishing Party”) which is, or whose Affiliates is, seeking to publish or publicly present scientific or technical data, results or other information with respect to the Product shall provide the other Party and the JDC with a copy of any proposed publication or presentation at least forty-five (45) days (or at least twenty (20) days in the case of abstracts or oral public presentations) prior to submission for publication or presentation so as to provide such other Party with an opportunity to recommend any changes it reasonably believes are necessary to continue to maintain such other Party’s Confidential Information in accordance with the requirements of this Agreement or to not jeopardize the patentability of any results or data.

 

(b)                                  If the non-Publishing Party notifies the Publishing Party that such publication or presentation, in the non-Publishing Party’s reasonable judgment, (i) discloses an invention for which the non-Publishing Party desires to seek patent protection, or (ii) contains any Confidential Information of the non-Publishing Party, or could be expected to have an adverse effect on the commercial value of any Confidential Information disclosed by the non-Publishing Party to the Publishing Party, the Publishing Party shall delete such Confidential Information from the proposed publication or presentation and shall further delay such publication or presentation for a period reasonably sufficient to permit the timely preparation and filing of a patent application(s) on any invention disclosed in such publication or presentation (but no more than ninety (90) days from the date of the non-Publishing Party’s notice thereof).

 

Section 10.6                              Use of Names.

 

(a)                                  Chiesi, its Affiliates and Third Party contractors shall not use the name “St. Jude Children’s Research Hospital” or any variation of that name, or any trademarks or logos belonging to St. Jude, or the names of any of St. Jude’s trustees, officers, faculty, student, employees, or agents, or any adaptation of such names, or any term of the St. Jude Agreement in any promotional material or other public announcement or disclosure or in connection with the Commercialization of the Product, without the prior written approval of St. Jude; except (i) in annual reports or as part of required regulatory or financial disclosures to the FDA, SEC or other US federal or foreign agencies; and (ii) where otherwise required by Applicable Laws, provided that, Chiesi shall notify St. Jude in advance of any disclosure to be made under these exceptions.

 

(b)                                  Chiesi, its Affiliates and Third Party contractors shall not state or imply that the PHS Agreements are an endorsement by the US government, PHS, any other US government organizational unit, or any US government employee. Additionally, Chiesi and its Affiliates shall not use the names of NIH, FDA, PHS, or HHS or the US government or their employees in any advertising, promotional, or sales literature without the prior written approval of PHS.

 

Section 10.7                              Term.  All obligations of confidentiality imposed under this Article X shall expire ten (10) years following termination or expiration of this Agreement, except to the extent any Existing Third Party License or Additional Rights Agreement extends such obligations; provided, however, that the receiving

 

39



 

Party shall maintain the confidentiality of any of the other Party’s trade secrets indefinitely until such trade secret is no longer a trade secret.

 

ARTICLE XI
REPRESENTATIONS, WARRANTIES AND COVENANTS

 

Section 11.1                              Representations and Warranties of Both Parties.  Each Party hereby represents and warrants to the other Party, as of the Effective Date, that:

 

(a)                                  such Party is duly organized, validly existing and in good standing under the Applicable Laws of the jurisdiction of its incorporation and has full corporate power and authority to enter into this Agreement and to carry out the provisions hereof;

 

(b)                                  such Party has taken all necessary action on its part to authorize the execution and delivery of this Agreement and the performance of its obligations hereunder;

 

(c)                                   this Agreement has been duly executed and delivered on behalf of such Party, and constitutes a legal, valid, binding obligation, enforceable against it in accordance with the terms hereof; and

 

(d)                                  the execution, delivery and performance of this Agreement by such Party does not conflict with any agreement, instrument or binding understanding, oral or written, to which it is a party or by which it is bound, nor violate any Applicable Law of any court, governmental body or administrative or other agency having jurisdiction over such Party.

 

Section 11.2                              Representations and Warranties of uniQure.  uniQure hereby represents and warrants to Chiesi, as of the Effective Date that:

 

(a)                                  Exhibit E attached hereto is a complete and correct list of all Licensed Patents that claim the composition of matter, or method of use or manufacture, of the Product and are Controlled by uniQure and for which uniQure controls Patent Prosecution as of the Effective Date;

 

(b)                                  uniQure Controls the Licensed Technology and has the full right, power and authority to grant all rights and licenses to Chiesi with respect to the Licensed Technology under this Agreement;

 

(c)                                   To uniQure’s knowledge, it has not (i) employed or used any contractor or consultant that employs any individual or entity debarred by the FDA (or subject to a similar sanction of EMA), or (ii) employed any individual or entity that is the subject of an FDA debarment investigation or proceeding (or similar proceeding of EMA), in each of clauses (i) and (ii), in the conduct of development activities directed to the Product;

 

(d)                                  To uniQure’s knowledge, the Development and Commercialization of the Product in the Territory, as anticipated hereunder, does not infringe upon any intellectual property rights of any Third Party

 

40



 

(e)                                   uniQure has not received any written allegation from a Third Party that any of the issued Licensed Patents is invalid or unenforceable and, except as disclosed in Exhibit H, to uniQure’s knowledge, none of such Licensed Patents is infringed by any Third Party;

 

(f)                                    uniQure has not received, with respect to the Product as Developed by uniQure, any written notice from a Third Party claiming infringement or misappropriation of any Patent Right or any Know-How owned by such Third Party; and

 

(g)                                   uniQure has provided Chiesi with a complete and correct copy of each of the Existing Third Party Licenses.

 

Section 11.3                              Representation and Warranty of Chiesi.  Chiesi hereby represents and warrants to uniQure, as of the Effective Date, that Chiesi Controls the Chiesi Technology and has the full right, power and authority to grant all rights and licenses to uniQure under this Agreement.

 

Section 11.4                              Mutual Covenants.  Each Party hereby covenants to the other Party that:

 

(a)                                  All employees of such Party or its Affiliates working under this Agreement are and will be under the obligation to assign all right, title and interest in and to their inventions and discoveries arising in the performance of such work, whether or not patentable, to (i) such Party as the sole owner thereof or (ii) to one of such Party’s Affiliates as the sole owner thereof so that such Party Controls such inventions and discoveries;

 

(b)                                  To its knowledge, such Party will not, in the conduct of its activities under this Agreement, (i) employ or use any contractor or consultant that employs any individual or entity debarred by the FDA (or subject to a similar sanction of EMA), or (ii) employ any individual who or entity that is the subject of an FDA debarment investigation or proceeding (or similar proceeding of EMA), in each of clauses (i) and (ii) in the conduct of its activities under this Agreement;

 

(c)                                   Such Party shall perform its activities pursuant to this Agreement in compliance in all material respects with Applicable Laws; and

 

(d)                                  Neither Party shall, during the Term, grant any right or license to any Third Party relating to any of the intellectual property rights it Controls which would conflict with, or limit the scope of, any of the rights or licenses granted or to be granted to the other Party hereunder.

 

Section 11.5                              DISCLAIMER.  EXCEPT AS OTHERWISE EXPRESSLY SET FORTH IN THIS AGREEMENT, NEITHER PARTY MAKES ANY REPRESENTATION OR EXTENDS ANY WARRANTY OF ANY KIND, EITHER EXPRESS OR IMPLIED, INCLUDING ANY WARRANTY THAT ANY PATENT RIGHTS ARE VALID OR ENFORCEABLE, AND EXPRESSLY DISCLAIMS ALL IMPLIED WARRANTIES OF MERCHANTABILITY, FITNESS FOR A PARTICULAR PURPOSE AND NON-INFRINGEMENT. WITHOUT LIMITING THE GENERALITY OF THE FOREGOING, EACH PARTY DISCLAIMS ANY

 

41



 

WARRANTIES WITH REGARDS TO: (A) THE SUCCESS OF ANY STUDY OR CLINICAL TRIAL COMMENCED UNDER THIS AGREEMENT; (B) THE SAFETY, USEFULNESS FOR ANY PURPOSE OR NON-INFRINGEMENT OF ANY PRODUCT; OR (C) THE VALIDITY, ENFORCEABILITY OR NON-INFRINGEMENT OF ANY INTELLECTUAL PROPERTY RIGHTS OR TECHNOLOGY IT PROVIDES OR LICENSES TO THE OTHER PARTY UNDER THIS AGREEMENT.

 

ARTICLE XII
TERM AND TERMINATION

 

Section 12.1                              Term.

 

(a)                                  General.  This Agreement shall become effective as of the Effective Date and shall remain in force, on a country-by-country basis, for the longer of (i) twelve (12) years from the First Commercial Sale of the Product in the relevant country of the Territory; (ii) expiry of any regulatory exclusivity granted by any Marketing Authorization or any other Regulatory Approval in the relevant country of the Territory; or (iii) expiry of the last Valid Claim Covering the Product in the relevant country of the Territory. Unless terminated by a Party with three (3) months written notice to the other Party to the end of the above initial or any subsequent term, this Agreement shall automatically be renewed for successive five (5) year terms (the initial and each subsequent term, the “Term”).

 

(b)                                  Condition Precedent.  This Agreement, and any ancillary agreement concluded between the Parties in connection herewith, including the SDEA, and the Commercialization Agreement and the agreement regarding the equity investment of Chiesi in uniQure concluded on the date hereof, shall become effective once the Parties have received consent from PHS as the Third Party licensor to the subcontracting of the rights and licenses licensed by uniQure as licensee under the PHS Agreements to Chiesi. uniQure shall use Commercially Reasonable Efforts to obtain such consent on or prior to 30 June 2013. If, despite uniQure’s Commercially Reasonable Efforts, such consent has not been obtained from PHS by the end of 30 June 2013, this Agreement and all other agreements that are subject to the condition precedent pursuant to sentence 1 shall be deemed null and void as of the Effective Date, unless, prior to the end of such period, following a corresponding request of either Party, the Parties mutually agree in writing on an extension of such period. The Parties agree that (i) costs and expenses incurred in connection with the preparation and execution of this Agreement as well as obtaining of the aforementioned consent shall not be reimbursed, provided, however, that uniQure shall pay back to Chiesi any payments received in connection with this Agreement on or prior to 30 June 2013 (or such extended period mutually agreed between the Parties in accordance with the foregoing), and (ii) Sections 10.1, 10.2 and 10.7 shall apply mutatis mutandis .

 

Section 12.2                              Termination for Convenience.  Chiesi may terminate this Agreement for convenience upon six (6) months’ prior written notice to uniQure at any time during the Term, following the first six (6) months of the Agreement.

 

Section 12.3                              Termination for Material Breach.  Upon any material breach of this Agreement by either Party (in such capacity, the “Breaching Party”), the other

 

42



 

Party (in such capacity, the “Non-Breaching Party”) may terminate this Agreement by providing sixty (60) days’ prior written notice to the Breaching Party, specifying the material breach. The termination shall become effective at the end of the sixty (60) day period unless the Breaching Party cures such breach during such sixty (60) day period.

 

In the event that either Party is the Breaching Party or that either Party, its Affiliates or Third Party contractors breach any of the requirements under any Existing Third Party Licenses or Additional Rights Agreements, such Party’s cure period described in the preceding paragraph shall be eliminated or reduced to the extent necessary to prevent the breach from giving a licensor a right to terminate an Existing Third Party License or any Additional Rights Agreement or from causing the other Party to be in breach of its obligations under any Existing Third Party License or any Additional Rights Agreement.

 

Section 12.4                              Termination for Patent Challenge.  If either Party or any of its Affiliates or Third Party contractors challenges the validity, enforceability, patentability or scope of any claim included in any Patent, (any of the foregoing, a “Patent Challenge”), the other Party shall have the right to terminate this Agreement immediately upon written notice to such Party.

 

Section 12.5                              Effects of Termination by Chiesi for Convenience or by uniQure for Chiesi Uncured Breach or Patent Challenge.  Upon termination of this Agreement by Chiesi in its entirety pursuant to Section 12.2 (Termination for Convenience) or by uniQure pursuant to Section 12.3 (Termination for Material Breach) or pursuant to Section 12.4 (Termination for Patent Challenge):

 

(a)                                  All rights and licenses granted by uniQure to Chiesi shall terminate and revert to uniQure, and all rights and licenses granted by Chiesi to uniQure shall survive and become fully paid-up, irrevocable and perpetual;

 

(b)                                  Chiesi shall promptly provide to uniQure a fair and accurate description of the status of its Development Program activities through the effective date of termination;

 

(c)                                   Chiesi shall promptly assign to uniQure the entire right, title, and interest in and to, and transfer to uniQure all copies of, any Product Data in Chiesi’s or its Affiliates’ or Third Party contractors’ possession or control;

 

(d)                                  At uniQure’s option and upon uniQure’s request as to any or all of the following (in whole or in part), Chiesi (or its relevant Affiliate) shall promptly:

 

(i)                          assign or cause the assignment to uniQure of any and all applicable Third Party agreements for the Product, including agreements with contract research organizations and other agreements relating to the Development of the Product, in each case to the extent assignable; provided, however, that, to the extent such agreements are not specific to the Product, or are not assignable, Chiesi shall, at uniQure’s request, hold such agreements for the benefit of uniQure and Chiesi and its Affiliates shall take such actions as uniQure may reasonably request so as to provide uniQure with the benefits thereunder with respect to the Product;

 

43



 

(ii)                       Chiesi and its Affiliates shall waive any obligations of confidentiality, non-competition and exclusivity imposed on its Third Party service providers, in order to permit uniQure to negotiate agreements with such service providers to develop, import, export and use the Product; and

 

(iii)                    at uniQure’s request, on a clinical trial-by-clinical trial basis with respect to any on-going clinical trial with the Product being conducted by or under authority of Chiesi or its Affiliates as of the date of the termination notice, either:

 

(A)                    terminate any such clinical trial as of the date of the termination notice in a manner conforming to Applicable Laws and provide all related data to uniQure promptly following termination of such clinical trial,

 

(B)                    continue to conduct such clinical trial to completion, keeping uniQure fully informed of the status of such clinical trial, and provide all related data to uniQure promptly following its completion, or

 

(C)                    promptly transfer such clinical trial to uniQure or its designee and continue to conduct such clinical trial up to completion of such transfer, keeping uniQure fully informed of the status of such clinical trial, and provide all related data to uniQure promptly following completion of such transfer;

 

(e)                                   In accordance with uniQure’s request, Chiesi shall promptly return to uniQure, or promptly destroy and certify to uniQure in writing that it has destroyed, all materials and records in its possession or Control containing Confidential Information of uniQure, except for a single copy of such Confidential Information that may be retained confidentially for legal purposes only;

 

(f)                                    With respect to Chiesi Patents, (i) the provisions of Section 9.2(b), Section 9.3(b) and Section 9.5 shall apply to the Chiesi Patents with the respective roles of the Parties reversed, and (ii) if uniQure initiates suit pursuant to Section 9.3(b), uniQure may retain any damages, settlements, accounts of profits, or other financial compensation recovered from a Third Party based upon such suit;

 

(g)                                   uniQure shall have the sole right to conduct Patent Prosecution with respect to the Joint Patents, notwithstanding Section 9.2(c), and to enforce the Joint Patents, notwithstanding Section 9.3;

 

(h)                                  Neither Chiesi nor any of its Affiliates shall use or grant rights to any Affiliate or Third Party for Joint Know-How in relation to any Competing Product during the one (1) year following termination;

 

(i)                                      The restrictions on Chiesi and its Affiliates set forth in Section 6.1 shall survive for one (1) year following termination; and

 

(j)                                     Chiesi shall execute all documents and take all such further actions as may be reasonably requested by uniQure in order to give effect to the foregoing clauses (a) through (i).

 

44



 

Section 12.6                              Effects of Termination by Chiesi for uniQure Uncured Breach or Patent Challenge.  Upon termination of this Agreement by Chiesi pursuant to Section 12.3 (Termination for Material Breach) or pursuant to Section 12.4 (Termination for Patent Challenge), the consequences set forth in Section 12.5 shall apply, mutatis mutandis ; provided that, the Parties shall negotiate in good faith appropriate consideration payable by uniQure to Chiesi in connection therewith reflecting the stage to which the Parties have Developed the Product prior to such termination.

 

Section 12.7                              Upon expiration of the Term with respect to this Agreement pursuant to Section 12.1:

 

(a)                                  all rights, privileges and licenses granted hereunder to Chiesi shall become fully paid-up, irrevocable and perpetual;

 

(b)                                  all rights, privileges and licenses granted hereunder to uniQure shall become fully paid-up, irrevocable and perpetual;

 

(c)                                   at any time upon written request of the disclosing Party, unless expressly set forth otherwise in this Agreement, the receiving Party shall cease use of and return or at the disclosing Party’s request destroy all Confidential Information of the disclosing Party and all copies thereof except for a single copy of such Confidential Information that may be retained confidentially for legal purposes only.

 

Section 12.8                              Survival.

 

(a)                                  Upon expiration or termination of this Agreement for any reason, all rights and obligations of each Party shall terminate hereunder, except as expressly set forth in Section 12.5, Section 12.6, Section 12.7 or this Section 12.8; provided, however, that nothing in this Agreement shall be construed to release either Party from any obligations or liabilities that matured prior to the effective date of expiration or termination, or which are attributable to a period prior to such expiration or termination.

 

(b)                                  Notwithstanding anything in this Agreement to the contrary, the following provisions shall expressly survive any expiration or termination of this Agreement in accordance with their terms: Article I, Section 6.2, Section 7.3, Article VIII (in each case, to the extent any amounts are due but unpaid as of the effective date of expiration or termination or thereafter pursuant to Section 12.5); Section 9.1; Article X; Section 11.5; Section 12.5; Section 12.6; Section 12.7; Section 12.8; Article XIII; Article XIV; and Article XV.

 

(c)                                   Termination of this Agreement shall be in addition to, and shall not prejudice, the Parties’ remedies at law or in equity, including the Parties’ ability to receive legal damages or equitable relief with respect to any breach of this Agreement, regardless of whether or not such breach was the reason for the termination.

 

45



 

ARTICLE XIII
DISPUTE RESOLUTION

 

Section 13.1                              Resolution of Certain Disputes Other Than by Arbitration.

 

(a)                                  Either Party’s exercise of its right to terminate this Agreement for the other Party’s material breach in accordance with Section 12.3, to the extent necessary to prevent such breach (whether by the other Party or its Affiliate) or from giving a licensor a right to terminate an Existing Third Party License or an Additional Rights Agreement or from causing the first Party to be in breach of its obligations under any Existing Third Party License or any Additional Rights Agreement, (a “Non-Arbitrable Termination Dispute”) shall not be subject to the dispute resolution procedures of Article II or Section 13.2 prior to termination. The other Party shall not be entitled to injunctive relief to prevent or delay such termination, and shall only be entitled to monetary damages in the event that it thereafter disputes such termination pursuant to Section 13.2(b)(i) and the arbitrators determine that the first Party has not properly exercised its termination right hereunder.

 

(b)                                  Any dispute that could be resolved by the JSC under Section 2.1(e), if unresolved by the JSC, shall be finally resolved as set forth in Section 13.2.

 

Section 13.2                              Resolution of Other Disputes by Executive Officers and Arbitration.

 

(a)                                  With the exception of Non-Arbitrable Termination Disputes, in the event any dispute arises out of or in relation to or in connection with any of the Collaboration Agreements, including any issue relating to the interpretation or application of the Collaboration Agreements, the Parties shall use good faith efforts to resolve such dispute within thirty (30) days, through the JSC if the dispute is within the responsibilities of the JSC, or, if the dispute is not within the responsibilities of the JSC, through informal negotiations between the respective representatives of the Parties.  If the JSC or the applicable representatives are unable to resolve such dispute within such thirty (30) day period, the Parties shall refer such dispute to the Executive Officers for resolution. If a dispute is referred to the Executive Officers for resolution pursuant to the preceding sentence (or pursuant to Section 2.1(f)), the Executive Officers shall attempt in good faith to resolve such dispute within thirty (30) days.

 

(b)                                  If the Executive Officers are unable to resolve a given dispute referred to such Executive Officers pursuant to Section 13.2(a) (or pursuant to Section 2.1(f) (other than Non-Arbitrable Termination Disputes) within thirty (30) days following such referral of such dispute to such Executive Officers, either Party may have the dispute settled by binding arbitration in the manner described below:

 

(i)                          Arbitration Request.  If a Party intends to begin an arbitration to resolve a dispute arising under a Collaboration Agreement, such Party shall provide written notice (the “Arbitration Request”) to the other Party of such intention and the issues for resolution.

 

(ii)                       Additional Issues.  Within ten (10) days after the receipt of the Arbitration Request, the other Party may, by written notice, add additional issues for resolution.

 

46



 

(iii)                    Arbitration Location; Rules.  Except as expressly provided herein, the sole mechanism for resolution of any claim, dispute or controversy arising out of or in connection with or relating to any Collaboration Agreement or the breach or alleged breach thereof shall be binding arbitration by ICC in London, England, pursuant to ICC’s Arbitration Rules and Procedures, except as provided herein.

 

(iv)                   English Language.  All proceedings shall be held in English and a transcribed record prepared in English. Documents submitted in the arbitration, the originals of which are not in English, shall be submitted together with a reasonably complete and accurate English translation.

 

(v)                      Selection of Arbitrators.  The Parties shall each select one arbitrator within thirty (30) days after receipt of the Arbitration Request and the two (2) arbitrators so selected shall select by mutual agreement a third arbitrator within thirty (30) days after they have been selected as arbitrators. If all three (3) arbitrators have not been selected within sixty (60) days after receipt of the Arbitration Request or any extension of time that is mutually agreed on, ICC shall select such additional arbitrator(s) needed to complete the three (3) arbitrator panel within thirty (30) days thereafter. If the issues in dispute involve scientific or technical matters, any arbitrators chosen hereunder shall have educational training or experience sufficient to demonstrate a reasonable level of knowledge in the pharmaceutical and biotechnology fields.

 

(vi)                   Time Schedule.  Within thirty (30) days after initiation of arbitration, the Parties shall reach agreement upon and thereafter follow procedures directed at assuring that the arbitration will be concluded and the award rendered within no more than six (6) months after selection of the three (3) arbitrators. Failing such agreement, ICC will design, and the Parties will follow procedures, directed at meeting such a time schedule.

 

(vii)                Powers of Arbitrators.  The arbitrators:

 

(A)                    shall not have any power or authority to add to, alter, amend or modify the terms of any Collaboration Agreement;

 

(B)                    shall establish and enforce appropriate rules to allow reasonable discovery by the Parties and to ensure that the proceedings, including the decision, be kept confidential and that all Confidential Information of the Parties be kept confidential and be used for no purpose other than the arbitration (unless disclosure or use is otherwise expressly permitted by the applicable Collaboration Agreement);

 

(C)                    shall have the power to enforce specifically the applicable Collaboration Agreement and the terms and conditions thereof in addition to any other remedies at law or in equity; and

 

(D)                    shall issue all awards in writing.

 

(viii)             Costs; Exclusion from Award.  Awards rendered by the arbitrators shall not include costs of arbitration, attorneys’ fees or costs for expert and

 

47



 

other witnesses, with respect to which each Party shall bear its own costs and expenses, except that the Parties shall share equally the fees of the arbitrators.

 

(ix)                   Injunctive Relief.  Nothing in this Agreement (except Section 13.1(a)) shall be deemed as preventing either Party from seeking injunctive relief (or any other provisional remedy such as temporary restraining order, preliminary injunction or other interim equitable relief) from the arbitrators or from any court having jurisdiction over the Parties (and prior to or during any arbitration if necessary to protect the interests of such Party in avoiding irreparable harm or to preserve the status quo pending the arbitration proceeding) and the subject matter of the dispute as necessary to protect either Party’s name, Confidential Information, trade secrets, Know-How or any other proprietary right or otherwise to avoid irreparable harm. In particular, the Parties agree that any breach by a Party of its obligations under Section 6.1, Section 7.5(a) or Article X, or any claim by either Party contrary to Section 13.1(a), will cause irreparable harm to the other Party for which an award of monetary damages would be an inadequate remedy and, accordingly, that the other Party shall be entitled to injunctive relief enjoining such breach without the requirement to post a bond.

 

(x)                      Judgment.  Judgment on any award rendered by the arbitrators may be entered in any court of competent jurisdiction.

 

ARTICLE XIV
INDEMNIFICATION; INSURANCE

 

Section 14.1                              Indemnification by Chiesi.  Chiesi shall indemnify, defend and hold harmless uniQure and its Affiliates, and its and their respective directors, officers, employees and agents, from and against any and all liabilities, damages, losses, costs and expenses, including the reasonable fees of attorneys and other professional Third Parties (collectively, “Losses”), arising out of or resulting from any and all Third Party suits, claims, actions, proceedings or demands (“Claims”) to the extent based upon:

 

(a)                                  any breach of any representation, warranty or covenant made by, or any material obligation of, Chiesi under this Agreement;

 

(b)                                  Development Program activities conducted by or on behalf (other than by uniQure or any of its Affiliates, or a Third Party performing activities on their behalf) of Chiesi or its Affiliates; or

 

(c)                                   the gross negligence, recklessness or willful misconduct of Chiesi or its Affiliates and its or their respective directors, officers, employees and agents;

 

provided that Chiesi shall not be obligated pursuant to this Section 14.1 to the extent uniQure is required to indemnify Chiesi under Section 14.2 below.

 

Section 14.2                              Indemnification by uniQure.  uniQure shall indemnify, defend and hold harmless Chiesi and its Affiliates, and its and their respective directors, officers, employees and agents, from and against any and all Losses, arising out of or resulting from any and all Claims to the extent based upon:

 

48



 

(a)                                  any breach of any representation, warranty or covenant made by, or any material obligation of, uniQure under this Agreement;

 

(b)                                  Development Program activities conducted by or on behalf (other than by Chiesi or any of its Affiliates, or a Third Party performing activities on their behalf) of uniQure or its Affiliates;

 

(c)                                   the gross negligence, recklessness or willful misconduct of uniQure or its Affiliates and its or their respective directors, officers, employees and agents; or

 

(d)                                  Claims that the exercise of any rights or licenses granted to Chiesi and its Affiliates in accordance with this Agreement violates or infringes upon the Intellectual Property Rights of any Third Party;

 

provided that uniQure shall not be obligated pursuant to this Section 14.2 to the extent Chiesi is required to indemnify uniQure under Section 14.1above.

 

Section 14.3                              Procedure.

 

(a)                                  A Party entitled to indemnification under this Article XIV (an “Indemnified Party”) shall give prompt written notification to the Party from whom indemnification is sought (the “Indemnifying Party”) of the commencement of any Claim for which indemnification may be sought or, if earlier, upon the assertion of any such Claim by a Third Party (it being understood and agreed, however, that the failure by an Indemnified Party to give notice of a Claim as provided in this Section 14.3(a) shall not relieve the Indemnifying Party of its indemnification obligation under this Agreement except and only to the extent that such Indemnifying Party is actually damaged as a result of such failure to give notice).

 

(b)                                  Within fifteen (15) days after delivery of such notification, the Indemnifying Party may, upon written notice thereof to the Indemnified Party, assume control of the defense of such Claim with counsel reasonably satisfactory to the Indemnified Party.

 

(c)                                   If the Indemnifying Party does not assume control of such defense, the Indemnified Party shall control such defense and, without limiting the Indemnifying Party’s indemnification obligations, the Indemnifying Party shall reimburse the Indemnified Party for all reasonable costs and expenses, including reasonable attorney’s fees, incurred by the Indemnified Party in defending itself, within thirty (30) days after receipt of any invoice therefor from the Indemnified Party, such invoice to be issued no more often than quarterly.

 

(d)                                  The Party not controlling such defense may participate therein at its own expense; provided that, if the Indemnifying Party assumes control of such defense and the Indemnified Party in good faith concludes, based on advice from counsel, that the Indemnifying Party and the Indemnified Party have conflicting interests with respect to such Claim, the Indemnifying Party shall be responsible for the reasonable fees and expenses of counsel to the Indemnified Party in connection with its participation in the defense action.

 

49



 

(e)                                   The Party controlling such defense shall keep the other Party advised of the status of such Claim and the defense thereof and shall consider recommendations made by the other Party with respect thereto.

 

(f)                                    The Indemnified Party shall not agree to any settlement of any Claim without the prior written consent of the Indemnifying Party, which shall not be unreasonably withheld, delayed or conditioned. The Indemnifying Party shall not, without the prior written consent of the Indemnified Party, agree to any settlement of such Claim, or consent to any judgment in respect thereof, that does not include a complete and unconditional release of the Indemnified Party from all liability with respect thereto, that imposes any liability or obligation on the Indemnified Party or that acknowledges fault by the Indemnified Party.

 

Section 14.4                              Insurance.  Each Party shall procure and maintain insurance, including product liability insurance, adequate to cover its obligations hereunder and which are consistent with normal business practices of comparable companies with respect to similar obligations and liabilities, at all times during the Term. uniQure shall further procure and maintain, at uniQure’s cost, insurance adequate to cover its obligations under the St. Jude Agreements and Chiesi shall reasonably cooperate with uniQure in obtaining such insurance. It is understood that such insurance shall not be construed to create any limit of either Party’s obligations or liabilities with respect to its indemnification obligations hereunder. Each Party shall provide the other, upon request, with evidence of such insurance.

 

Section 14.5                              Limitation of Liability.  EXCEPT WITH RESPECT TO ANY BREACH BY A PARTY OF ITS OBLIGATIONS UNDER ARTICLE X, EXCEPT FOR ANY DAMAGES ARISING FROM A PARTY’S WILLFUL MISCONDUCT AND EXCEPT TO THE EXTENT A PARTY MAY BE REQUIRED TO INDEMNIFY THE OTHER PARTY UNDER THIS ARTICLE XIV WITH RESPECT TO THIRD PARTY CLAIMS, NEITHER PARTY SHALL BE LIABLE FOR ANY (AND EACH PARTY HEREBY DISCLAIMS ALL) SPECIAL, EXEMPLARY, CONSEQUENTIAL, PUNITIVE OR OTHER INDIRECT DAMAGES, INCLUDING LOST REVENUE AND LOST PROFITS, WHETHER BASED UPON WARRANTY, CONTRACT, TORT, STRICT LIABILITY OR OTHER LEGAL THEORY.

 

ARTICLE XV
MISCELLANEOUS

 

Section 15.1                              Change of Control.  Each Party agrees that, notwithstanding any provisions of this Agreement to the contrary, following the closing of a Change of Control of a Party (the “Acquired Party”), the other Party (the “Non-Acquired Party”) shall not obtain rights or access to the Patent Rights or Know-How of the Acquirer (as defined below) or of the Affiliates of such Acquirer (other than the Acquired Party and its Affiliates which exist immediately prior to the closing of such Change of Control (such Affiliates, the “Pre-Existing Affiliates”)); and the Acquirer and its Affiliates (other than the Acquired Party and its Pre-Existing Affiliates) shall not obtain rights or access to the Patent Rights or Know-How of the Non-Acquired Party or be bound by the restrictions set forth in Section 6.1; provided, however, that the Non-Acquired Party’s rights in all Patent Rights and Know-How of the Acquired Party and its Pre-Existing Affiliates, which Patent Rights and Know-How exist as of

 

50



 

the date of the closing of such Change of Control and are then licensed hereunder to the Non-Acquired Party, shall remain licensed to such Non-Acquired Party after the date of the closing of such Change of Control in accordance with and subject to the terms and conditions of this Agreement and shall not be affected in any manner by virtue of such Change of Control. “Acquirer” means, with respect to the Acquired Party, the Third Party that acquires the Control of such Acquired Party.

 

Section 15.2                              Governing Law.  The validity and interpretation of this Agreement shall be governed by the laws of England without regard to its conflicts of laws principles and to the express exclusion of the United Nations Conventions on Contracts for the International Sale of Goods (CISG).

 

Section 15.3                              Assignment.  Except as expressly provided herein, neither this Agreement nor any rights and obligations hereunder shall be assignable by a Party without the prior written consent of the other Party; provided, however, that a Party may assign this Agreement to any Affiliate or to any successor in interest by way of merger, acquisition or sale of all or substantially all of its assets to which this Agreement relates, provided that such successor agrees in writing to be bound by the terms of this Agreement as if it were the assigning Party. This Agreement shall be binding upon the successors and permitted assigns of the Parties. Any assignment not in accordance with this Section 15.3 shall be void.

 

Section 15.4                              Entire Agreement; Amendments.  This Agreement and the attachments hereto contain the entire understanding and agreement of the Parties with respect to the subject matter hereof and cancel and supersede any and all prior negotiations, correspondence, understandings and agreements between the Parties, whether oral or written, regarding such subject matter, including the Memorandum of Understanding dated 21 December 2012 and the Confidentiality Agreement, but expressly excluding the Commercialization Agreement. Except for the rights expressly conferred on the JSC, JDC or JCC, this Agreement cannot be modified except by a written document bearing the signatures of both Parties. The same applies to any waiver of this written form requirement.

 

Section 15.5                              Notices.  Other than as expressly specified in this Agreement, all notices and consents required to be provided hereunder shall be in writing and provided by hand, by recorded delivery mail (return receipt requested), by facsimile, or by recognized overnight courier service to the other Party at its address or facsimile number shown below or such other address or facsimile number notified by such other Party from time to time.

 

If to uniQure, addressed to:

 

uniQure Biopharma B.V.

P.O. Box 22506

1100 DA Amsterdam

The Netherlands

Attention: CEO

Fax: +31 20 566 9272

 

If to Chiesi, addressed to:

 

51



 

Chiesi Farmaceutici S.p.A.

Via Palermo, 26/A

43122 Parma

Italy

Attention: CEO

Copy to: Corporate Development, Head and General Counsel

Fax: +39 0521 774468

 

Section 15.6                              Exports.  The Parties acknowledge that the export of technical data, materials or products is subject to the exporting Party receiving any necessary export licenses and that the Parties cannot be responsible for any delays attributable to export controls that are beyond the reasonable control of either Party. Chiesi and uniQure agree not to export or re-export, directly or indirectly, any Product (or any associated products, information, items, articles, computer software, media, technical data, the direct product of such data, samples or equipment received or generated under this Agreement) in violation of any Applicable Laws that may be applicable. Chiesi and uniQure agree to obtain similar covenants from their Affiliates and Third Party contractors with respect to the subject matter of this Section 15.6, to the extent applicable.

 

Section 15.7                              Force Majeure.  Neither Party shall lose any rights hereunder or be liable to the other Party for damages or losses on account of failure of performance by the defaulting Party if the failure is occasioned by war, civil insurrection, strike, fire, Act of God, earthquake, tempest, flood, epidemic, blackout, lockout, embargo, governmental acts or orders or restrictions, delays in delivery and non-supply by exclusive suppliers, where such delay or non-supply occurs as a result of such force majeure, or any other reason where failure to perform is beyond the reasonable control of such Party and such failure to perform is not caused by the negligence, intentional conduct or misconduct of the non-performing Party and such Party has exerted all Commercially Reasonable Efforts to avoid or remedy such force majeure event; provided, however, that in no event shall a Party be required to settle any labor dispute or disturbance.

 

Section 15.8                              Performance by Affiliates, Sub-distributors or Third Party Contractors.  To the extent that this Agreement imposes obligations on or permits the exercise of rights by Affiliates, Sub-distributors or Third Party contractors of a Party, such Party shall cause such Party’s Affiliates, Sub-distributors or Third Party contractors to perform such obligations (and all related obligations) and shall remain responsible for any breach of such obligations and for the exercise of rights by such Party’s Affiliates, Sub-distributors or Third Party Contractors.

 

Section 15.9                              Independent Contractors.  It is understood and agreed that the relationship between the Parties hereunder is that of independent contractors and that nothing in this Agreement shall be construed as authorization for either uniQure or Chiesi to act for, bind or commit the other in any way. Each Alliance Manager shall be considered the employee of Chiesi or uniQure, as the case may be, and shall not be deemed to be an employee of the other Party.

 

Section 15.10                       Costs.  Except as expressly provided in this Agreement or as separately agreed upon in writing between the Parties, each Party shall bear its own

 

52



 

costs incurred in connection with the implementation of the obligations under this Agreement.

 

Section 15.11                       Construction.  Each Party agrees that this Agreement shall be interpreted without regard to any presumption or rule requiring construction against the Party causing this Agreement to be drafted.

 

Section 15.12                       English Language.  This Agreement was prepared and is established in the English language; any translation thereof shall be deemed for convenience only and shall never prevail against the original English version. All reports, notices and communications to be exchanged under this Agreement shall be in the English language; provided, however, that, neither Party shall be under any obligation to translate into English any document originally established and existing in another language, for the sole purpose of communicating such document to the other Party, it being agreed that such documents will be provided on an as-is basis.

 

Section 15.13                       No Implied Waivers; Rights Cumulative.  No failure on the part of a Party to exercise, and no delay in exercising, any right, power, remedy or privilege under this Agreement, or provided by statute or at law or in equity or otherwise, shall impair, prejudice or constitute a waiver of any such right, power, remedy or privilege or be construed as a waiver of any breach of this Agreement or as an acquiescence therein, nor shall any single or partial waiver of any such right, power, remedy or privilege preclude any other or further exercise thereof or the exercise of any other right, power, remedy or privilege. Any Party may waive its rights hereunder in a writing signed by such Party.

 

Section 15.14                       Severability.  If, under Applicable Law, any provision of this Agreement is held to be invalid or unenforceable, or otherwise directly or indirectly affects the validity of any other material provision(s) of this Agreement (such invalid or unenforceable provision, a “Severed Clause”), this Agreement shall endure except for the Severed Clause. The Parties shall consult one another and use reasonable efforts to agree upon a valid and enforceable provision that is a reasonable substitute for the Severed Clause in view of the objectives contemplated by the Parties when entering into the Agreement and the general balance of the respective interests of the Parties as initially intended under the Agreement.

 

Section 15.15                       Counterparts.  This Agreement may be executed in two (2) or more counterparts, each of which will be deemed an original, but all of which together will constitute one and the same instrument. A pdf file of this Agreement contained in an email, including the signed signature pages hereto, will be deemed to be an original.

 

[ Signature Page Follows ]

 

53



 

IN WITNESS WHEREOF, the Parties have executed this Co-Development and License Agreement as of the Effective Date.

 

UNIQURE BIOPHARMA B.V.

 

UNIQURE BIOPHARMA B.V.

 

 

 

 

 

 

 

 

 

By:

/s/ Piers Morgan

 

By:

/s/ Hans Preusting

 

Name: Mr. Piers Morgan

 

 

Name: Mr. Hans Preusting

 

Title: Chief Financial Officer

 

 

Title: Business Development, Vice President

 

 

 

 

 

 

CHIESI FARMACEUTICI S.p.A.

 

CHIESI FARMACEUTICI S.p.A.

 

 

 

 

 

 

 

 

 

By:

/s/ Alberto Chiesi

 

By:

/s/ Ugo Di Francsco

 

Name: Mr. Alberto Chiesi

 

 

Name: Mr. Ugo Di Francesco

 

Title: President

 

 

Title: CEO

 

54



 

EXHIBIT A

 

Initial Development Plan and Budget

 

AMT-060 Key Development Activities to MAA

 

Foregrounds and assumptions to the Development Plan

 

·                   The present document is a high level overview and not an exhaustive list of all the work and activities involved in the development of AMT060.

 

·                   The timelines have been developed based on current knowledge and understanding of what is necessary in order to obtain marketing authorization.

 

·                   Plans and decisions may change depending on emerging data and information, according to the decisions taken by the JDC.

 

·                   The present plan does not include some tasks/ activities that are already foreseen, but which cannot be planned at the present stage due to missing information or for which decision making is expected to occur later on in development. Those activities include:

 

1.               The paediatric development, including pre-clinical experiments, Paediatric Investigation Plan (PIP) generation and approval (including appropriate regulatory interactions), and paediatric clinical trials. The timing for generation and submission of the PIP to the EMA will be decided by the JDC based on relevant experimental data and regulatory strategy.

 

2.               Extension of indication to moderately severe  patients. The decision on whether to extend the target indication to patients with moderately severe hemophilia (i.e. with spontaneous factor IX circulating activity 1-5% of normal) will be taken by the JDC mainly based on data from the Phase I study. The clinical and regulatory development strategy for this additional population will also be agreed at JDC level. NOTE: Severe = less than 1%; Moderate or moderately severe = 1-5%; mild = 5-50%.  The current plan is targeting the severe population only.

 

3.               The strategy for potential re-administration (including the decision whether to pursue re-administration and the clinical/ regulatory development plan for the indication) will be discussed and agreed by the JDC.

 

Tables 1 and 2 below highlight key activities in the AMT-060 development programme.

 

A- 1



 

Table 1: Key activities needed to deliver Phase I

 

uniQure Function

 

Activity

 

Start
Date (1)

 

Completion
Date (2)

 

Status

 

Regulatory affairs

 

IMPD finalization*

 

Dec 2013

 

Dec 2013

 

Planned

 

Non-clinical

 

NHPGLPtox. & safety study

 

Sep 2012

 

Aug 2013

 

Ongoing

 

 

 

Mouse GLPtox, safety &biodistribution study

 

Jan 2013

 

Dec 2013

 

Ongoing

 

Process Development

 

Tech transfer to MF

 

Dec 2012

 

Mar 2013

 

Close to completion

 

 

 

Viral clearance

 

Mar 2013

 

Jun 2013

 

Ongoing

 

Assay Development

 

Testing, specification & readiness assay package

 

Nov 2012

 

Jun 2013

 

Ongoing

 

 

 

Stability of tox and clinical batch

 

Nov 2012

 

Jun 2016

 

Ongoing

 

 

 

Clinical assay development

 

Jul 2012

 

Mar 2014

 

Ongoing

 

Quality control

 

MSV/ WSV testing

 

Aug 2012

 

Jun 2013

 

Ongoing

 

 

 

Phase I batch analysis & release testing*

 

Jul 2013

 

Nov 2013

 

Planned

 

Manufacturing

 

Phase I batch production

 

Jul 2013

 

Aug 2013

 

Planned

 

Quality Assurance

 

Phase I batch review and QP release

 

Jul 2013

 

Nov 2013

 

Planned

 

Clinical

 

Clinical Trial Application

 

Jan 2014

 

Jan 2014

 

Planned

 

 

 

Phase I start/ end

 

Mar – May 2014

 

3Q2015 (3)

 

Planned

 

 


(1) “Start date” means the date when the activity is planned to start. For clinical studies, “start date” means the first patient enrolled in the study (i.e. First-Patient-First-Visit, FPFV). For regulatory activities, it means the start of the window period in which the activity will occur.

 

(2) “Completion date” means the date by which the activity is planned to end. For clinical studies, “completion date” means the completion of the Clinical Study Report (i.e. CSR). For regulatory activities, it means the end of the window period in which the activity will occur.

 

(3) The planned “completion date” for the Phase I study assumes: (1) a 12-week run-in period before treatment of each patient; (2) the enrollment of 4 cohorts of patients with a 2-week period to complete AMT-060 administration within each cohort; (3) a 4-week interval between consecutive cohorts (CHMP recommended an 8 week interval, however it commented that it was in any case difficult to come up with a reasonable advice); and (4) a 24-week follow-up period to assess the safety/ efficacy end-points; for a total study duration from FPFV to Last-Patient-Last-Visit (LPLV) of 52 weeks (compared to the 68 weeks foreseen if the 8-week between-cohort period will be imposed by Heath Authorities and/ or Ethical Committees).

 

* Indicates critical path activities

 

A- 2



 

Table 2: Key activities needed to deliver Phase II/III

 

uniQure Function

 

Activity

 

Start
Date (1)

 

Completion
Date (2)

 

Status

Regulatory affairs

 

EMA Scientific Advice on quality meeting request*

 

Sep 2013

 

Oct 2013

 

Planned

 

 

EMA Phase II/ III protocol advice meeting request*

 

Oct 2014

 

Nov 2014

 

Planned

 

 

MAA submission

 

Jun 2017

 

Jun 2017

 

Planned

Non clinical

 

In vitro/ in vivo scale-up comparability studies

 

May 2014

 

Aug 2014

 

Planned

Process Development

 

Scale-up feasibility*

 

Apr 2013

 

Dec 2013

 

Planned (already initiated strategy approach & work-package outline)

 

 

Go/ No Go Decision to purchase 250L; [Go/ No Go Decision for scale-out]

 

Dec 2013

 

Jan 2014

 

Planned

 

 

Scale up process development (incl. transfer to MF)*

 

Jan 2014

 

Apr 2014

 

Planned

 

 

Process & analytical validation

 

Apr 2014

 

Sep 2014

 

Planned

Assay Development

 

Release assay validation and transfer to QC

 

Oct 2013

 

Jan 2014

 

Planned

 

 

Stability on phase II/III batch material

 

Jan 2015

 

Dec 2017

 

Planned

 

 

Molecular characterization of NHP Fix vshFIX

 

TBC

 

Apr 2014

 

To be planned

Quality control

 

Phase II/III batch analysis & release testing

 

Oct 2014

 

Mar 2015

 

Planned

Manufacturing

 

Phase II/III batch production

 

Oct 2014

 

Dec 2014

 

Planned

Quality Assurance

 

Phase II/III batch review and QP release

 

Oct 2014

 

May 2015

 

Planned

Clinical

 

Clinical Trial Application

 

Jun 2015

 

Jun 2015

 

Planned

 

 

Phase II/III start/ end

 

Jul - Sept 2015

 

2Q2017

 

Planned

 


(1), (2), and * As for Table 1.

 

A- 3



 

FTE requirements over the next 2 year

 

Table 3 shows the maximum FTE required by UniQure per year over the next 2 years (i.e. 2013 & 2014).  This timeline captures all activities necessary to support and initiate the phase I clinical trial and make significant progress with regards to preparing for the Phase II/III.

 

·                   These numbers include current staff as well as upcoming hires in PD (x1 technical post) and QC (x2 scientist/ technical post).  The new hires are to support all work in these Functions, not just AMT-060.

 

·                   The figures also include the estimated numbers for scale-up activities (i.e. x2,5 PD, x2 AD, x1.5 MF, x0.5 QA).

 

Table 4 indicates the maximum number of FTEs requirements for the time period 2015 to 2017 and Table 5 shows AMT-060 development costs (FTEs and external costs) for the 5 year perios, 2013 to 2017.

 

NOTE: FTEs and costs beyond 2017 to be determined and agreed by the JDC.

 

Table 3: Maximum FTE number per year per Function

 

uniQure
Function*

 

FTE AV.
2013

 

FTE AV.
2014

 

RA

 

 

1.3

 

0.5

 

NC

 

 

0.8

 

0.6

 

R&D

Res

 

 

0.4

 

0.6

 

AD

AD

 

8.3

 

2.9

 

BioAD

 

0.5

 

0.7

 

AD-S

 

0.3

 

0.3

 

PD

 

 

2.5

 

2.5

 

QC

 

 

2.9

 

3.0

 

QA

 

 

3.5

 

2.1

 

MF

MF

 

 

2.5

 

3.3

 

OPS

 

 

0.1

 

0.1

 

CD

 

1.6

 

2.6

 

PM

 

1.0

 

1.0

 

AV. TOTAL

 

25.4

 

20.2

 

 


*RA: Regulatory Affairs; NC: Non clinical; Res: Research; AD: Analytical Development; BioAD: Bio-Analytical Development; AD-S: Analytical Development Stability; PD: Process Development; QC: Quality Control; QA: Quality Assurance; MF: Manufacturing; OPS: Operations; CD: Clinical Development; PM: Project Management

 

A- 4



 

Table 4: Maximum FTE requirement 2015 - 2017:

 

Year

 

2015

 

2016

 

2017

 

Total FTE

 

25

 

25

 

10

 

Clinical

 

2

 

2

 

2

 

R&D

 

10,5

 

10,5

 

4

 

Manufacturing

 

3

 

3

 

0,5

 

QC

 

4

 

4

 

1

 

Non-clinical

 

1

 

1

 

0,3

 

QA

 

1,5

 

1,5

 

0,2

 

RA

 

2

 

2

 

1

 

Project Mgmt

 

1

 

1

 

1

 

 

Table 5: AMT-060 total budget and costs 2013 — 2017

 

 

 

2013

 

2014

 

2015

 

2016

 

2017

 

5 year
TOTAL

 

FTE (Number)

 

25,4

 

20,2

 

25

 

25

 

10

 

105,6

 

FTE rate (€/year)

 

250.000

 

250.000

 

250.000

 

250.000

 

250.000

 

 

 

Total FTE cost* (€)

 

6.350.000

 

5.050.000

 

6.250.000

 

6.250.000

 

2.500.000

 

26.400.000

 

External costs (€) [w/o cost for GMP batches]

 

2.798.000

 

2.295.000

 

3.590.000

 

2.310.000

 

1.410.000

 

12.403.000

 

GMP batches cost (€) [manufacturing]

 

 

 

 

 

 

 

 

 

 

 

(Total No. of Batches) x €91.000

 

Scale-up capital investment (€)

 

1.000.000#

 

 

 

 

 

1.000.000

 

TOTAL BUDGET (€)

 

10.148.000

 

7.345.000

 

9.840.000

 

8.560.000

 

3.910.000

 

39.803.000+ (No of Batches x €91.000 )

 

 


*Total FTE cost/year = (FTE number for that year) x (FTE rate per year)
# It is possible that some of this cost will be carried over in 2014

 

NOTE: all external costs estimates and costs allocation per year are based on current knowledge and planning.

 

A- 5



 

Outline of proposed clinical studies

 

Phase I study

 

Study

Phase I, multicentre, open label, prospective, interventional, single dose, dose-escalation clinical trial to investigate the safety and tolerability of AAV5-hFIXco, an adeno-associated viral vector containing a codon-optimised human factor IX gene in severe Haemophilia B (Study code: AMT-060-001)

 

 

Objectives and end-points

Primary objective:

 

 

 

To assess the safety of systemic and determine the MTD of the gene therapy vector AAV5-hFIXco for the treatment of the Severe Haemophila B, registering and evaluating the occurrence of adverse events and serious adverse events at the dose identified.

 

 

 

Main secondary objectives:

 

 

 

·                   To estimate the appropriate dose required to achieve stable expression of hFIX at or above 3% of normal

 

·                   To evaluate kinetics (dose-related duration and magnitude) of expression

 

·                   To describe the immune response to hFIX transgene product

 

·                   To describe the immune response to the AAV5 capsid proteins

 

·                   To assess viral shedding in various body fluids (including semen)

 

·                   Assess the occurrence of FIX inhibitors

 

·                   Evaluate coagulation parameters

 

·                   Assess need for FIX concomitant treatment

 

 

Study design

·                   Open label, non-randomized, prospective, interventional, single dose, dose-escalation phase I clinical trial.

 

·                   12 weeks of run-in period

 

·                   24 week monitoring period after administration

 

·                   Patients will be followed for safety and therapeutic response at intervals of 24 weeks for 5 years.

 

 

Study population

·                   Twelve male adults

(key eligibility criteria)

·                   > 18 year old to < 35 year old

 

·                   Genetically confirmed Haemophilia B (resulting from the missense mutation in the coagulation factor IX gene)

 

·                   Phenotypically defined as Severe disease ( < 1% of normal plasma FIX levels)

 

·                   Have had a total of 20 or more exposure days of treatment with FIX protein concentrate and an average of 3 bleeding episodes per year requiring FIX replacement therapy

 

·                   No evidence of active hepatitis or relative hepatic insufficiency

 

·                   No presence of anti-hFIX antibodies (“inhibitors”)

 

·                   No evidence of neutralizing antibodies (Nabs) against AAV5 according to pre-defined criteria

 

A- 6



 

 

·                   Normal range of liver enzymes plasma levels

 

 

Treatments

Cohort A: 5x10e11 gc/kg

 

Cohort B: 2x10e12 gc/kg

 

Cohort C: 6x10e12 gc/kg

 

Cohort D: 1.2x10e13 gc/kg

 

 

Duration

The trial will last 37 weeks for each patient

 

 

Sample size

12 patients

 

Phase II/ III: No definitive plans have been agreed yet.  It is envisaged that it will be a single dose confirmatory trial where the study population (28 patients with severe haemophilia) and the outcomes (efficacy endpoints — clinical and biochemical) assessed will be based on those as for phase I.  It is assumed that the trial will take 18 months from start to completion.

 

A- 7



 

EXHIBIT B

 

Certain Requirements under PHS Agreements

 

Each capitalized term used but not defined in this Exhibit B shall have the meaning ascribed to it in the applicable PHS Agreement.

 

PHS 2011 Requirements

 

4.2                                Licensee agrees that any sublicenses granted by it shall provide that the obligations to PHS of Paragraphs 5.1-5.4, 8.1, 10.1, 10.2, 12.5, and 13.8-13.10 of this Agreement shall be binding upon the sub-licensee as if it were a party to this Agreement. Licensee further agrees to attach copies of these Paragraphs to all sublicense agreements.

 

5.1                                (a)           PHS reserves on behalf of the Government an irrevocable, nonexclusive, nontransferable, royalty-free license for the practice of all inventions licensed under the Licensed Patent Rights throughout the world by or on behalf of the Government and on behalf of any foreign government or international organization pursuant to any existing or future treaty or agreement to which the Government is a signatory. Prior to the First Commercial Sale, Licensee agrees to provide PHS with reasonable quantities of Licensed Products or materials made through the Licensed Processes for PHS research use; and

 

(b)           In the event that the Licensed Patent Rights are Subject Inventions made under a Cooperative Research and Development Agreement (“CRADA”), Licensee grants to the Government, pursuant to 15 U.S.C. §3710a(b)(1)(A), a nonexclusive, nontransferable, irrevocable, paid-up license to practice Licensed Patent Rights or have Licensed Patent Rights practiced throughout the world by or on behalf of the Government. In the exercise of this license, the Government shall not publicly disclose trade secrets or commercial or financial information that is privileged or confidential within the meaning of 5 U.S.C. §552(b)(4) or which would be considered as such if it had been obtained from a non-Federal party. Prior to the First Commercial Sale, Licensee agrees to provide PHS reasonable quantities of Licensed Products or materials made through the Licensed Processes for PHS research use.

 

5.2                                Licensee agrees that products used or sold in the United States embodying Licensed Products or produced through use of Licensed Processes shall be manufactured substantially in the United States, unless a written waiver is obtained in advance from PHS.

 

5.3                                Licensee acknowledges that PHS may enter into future CRADAs under the Federal Technology Transfer Act of 1986 that relate to the subject matter of this Agreement. Licensee agrees not to unreasonably deny requests for a Research License from future collaborators with PHS when acquiring these rights is necessary in order to make a CRADA project feasible. Licensee may request an opportunity to join as a party to the proposed CRADA.

 

5.4                                (a)           In addition to the reserved license of Paragraph 5.1, PHS reserves the right to grant Research Licenses directly or to require Licensee to grant Research Licenses on reasonable terms. The purpose of these Research Licenses is to encourage basic

 

B- 1



 

research, whether conducted at an academic or corporate facility. In order to safeguard the Licensed Patent Rights, however, PHS shall consult with Licensee before granting to commercial entities a Research License or providing to them research samples of materials made through the Licensed Processes; and

 

(b)           In exceptional circumstances, and in the event that Licensed Patent Rights are Subject Inventions made under a CRADA, the Government, pursuant to 15 U.S.C. §3710a(b)(1)(B), retains the right to require the Licensee to grant to a responsible applicant a nonexclusive, partially exclusive, or exclusive sublicense to use the Licensed Patent Rights in the Licensed Field of Use on terms that are reasonable under the circumstances, or if Licensee fails to grant this license, the Government retains the right to grant the license itself. The exercise of these rights by the Government shall only be in exceptional circumstances and only if the Government determines:

 

(i)                                      the action is necessary to meet health or safety needs that are not reasonably satisfied by Licensee;

 

(ii)                                   the action is necessary to meet requirements for public use specified by Federal regulations, and these requirements are not reasonably satisfied by the Licensee; or

 

(iii)                                the Licensee has failed to comply with an agreement containing provisions described in 15 U.S.C. §3710a(c)(4)(B); and

 

(c)           The determination made by the Government under this Paragraph 5.4 is subject to administrative appeal and judicial review under 35 U.S.C. §203(b).

 

10.1                         Licensee shall use its reasonable commercial efforts to bring the Licensed Products and Licensed Processes to Practical Application. “Reasonable commercial efforts” for the purposes of this provision shall include adherence to the Commercial Development Plan in Appendix E and performance of the Benchmarks in Appendix D. The efforts of a sub-licensee shall be considered the efforts of Licensee.

 

10.2                         Upon the First Commercial Sale, until the expiration or termination of this Agreement, Licensee shall use its reasonable commercial efforts to make Licensed Products and Licensed Processes reasonably accessible to the United States public.

 

12.5                         Licensee shall indemnify and hold PHS, its employees, students, fellows, agents, and consultants harmless from and against all liability, demands, damages, expenses, and losses, including but not limited to death, personal injury, illness, or property damage in connection with or arising out of:

 

(a)                                  the use by or on behalf of Licensee, its sub-licensees, directors, employees, or third parties of any Licensed Patent Rights; or

 

(b)                                  the design, manufacture, distribution, or use of any Licensed Products, Licensed Processes or materials by Licensee, or other products or processes developed in connection with or arising out of the Licensed Patent Rights.

 

13.8                         PHS reserves the right according to 35 U.S.C. §209(d)(3) to terminate or modify this Agreement if it is determined that this action is necessary to meet the requirements for

 

B- 2



 

public use specified by federal regulations issued after the date of the license and these requirements are not reasonably satisfied by Licensee.

 

13.9                         Within thirty (30) days of receipt of written notice of PHS’ unilateral decision to modify or terminate this Agreement, Licensee may, consistent with the provisions of 37 C.F.R. §404.11, appeal the decision by written submission to the designated PHS official. The decision of the designated PHS official shall be the final agency decision. Licensee may thereafter exercise any and all administrative or judicial remedies that may be available.

 

13.10                  Within ninety (90) days of expiration or termination of this Agreement under this Article 13, a final report shall be submitted by Licensee. Any royalty payments, including those incurred but not yet paid (such as the full minimum annual royalty), and those related to patent expense, due to PHS shall become immediately due and payable upon termination or expiration. If terminated under this Article 13, sub-licensees may elect to convert their sublicenses to direct licenses with PHS pursuant to Paragraph 4.3.Unless otherwise specifically provided for under this Agreement, upon termination or expiration of this Agreement, Licensee shall return all Licensed Products or other materials included within the Licensed Patent Rights to PHS or provide PHS with certification of the destruction thereof. Licensee may not be granted additional PHS licenses if the final reporting requirement is not fulfilled.

 

PHS 2007 Requirements

 

4.2                                Licensee agrees that any sublicenses granted by it shall provide that the obligations to PHS of Paragraphs 5.1, 5.2, 8.1, 10.1, 10.2, 12.5 and 13.6-13.8 of this Agreement shall be binding upon the sub-licensee as if it were a party to this Agreement. Licensee further agrees to attach copies of these Paragraphs to all sublicense Agreements.

 

5.1                                Prior to the First Commercial Sale, Licensee agrees to provide PHS with reasonable quantities of Licensed Products or New Products made through the Licensed Processes or Supplied Materials solely for PHS research use, if requested in writing.

 

5.2                                Licensee agrees that products used or sold in the United States embodying Licensed Products or New Products or produced through use of Licensed Processes shall be manufactured substantially in the United States, unless a written waiver is obtained in advance from PHS.

 

8.1                                Licensee agrees to keep accurate and correct records of Licensed Products or New Products made, used, sold, or imported and Licensed Processes practiced under this Agreement appropriate to determine the amount of royalties due PHS. These records shall be retained for at least five (5) years following a given reporting period and shall be available during normal business hours for inspection, at the expense of PHS, by an accountant or other designated auditor selected by PHS for the sole purpose of verifying reports and royalty payments hereunder. The accountant or auditor shall only disclose to PHS information relating to the accuracy of reports and royalty payments made under this Agreement. If an inspection shows an underreporting or underpayment in excess of five percent (5%) for any twelve (12) month period, then Licensee shall reimburse PHS for the cost of the inspection at the time Licensee pays the unreported royalties, including any additional royalties as required by Paragraph

 

B- 3



 

9.8, All royalty payments required under this Paragraph shall be due within thirty (30) days of the date PHS provides Licensee notice of the payment due.

 

10.1                         Licensee shall use its reasonable commercial efforts to bring the Licensed Products or New Products and Licensed Processes to Practical Application. “Reasonable commercial efforts” for the purposes of this provision shall include adherence to the Commercial Development Plan in Appendix E and performance of the Benchmarks in Appendix D. The efforts of a sublicense shall be considered the efforts of Licensee.

 

10.2                         Upon the First Commercial Sale, until the expiration or termination of this Agreement, Licensee shall use its reasonable commercial efforts to make Licensed Products or New Products and Licensed Processes reasonably accessible to the United States public.

 

12.5                         Licensee shall indemnify and hold PHS, its employees, students, fellows, agents, and consultants harmless from and against all liability, demands, damages, expenses, and losses, including but not limited to death, personal injury, illness, or property damage in connection with or arising out of:

 

(a)                                  the use by or on behalf of Licensee, its sub-licensees, its directors, employees, or third parties of any Licensed Patent Rights or Supplied Materials; or

 

(b)                                  the design, manufacture, distribution, or use of any Licensed Products, Licensed Processes or New Products by Licensee, or other products or processes developed in connection with or arising out of the Licensed Patent Rights. Licensee agrees to maintain a liability insurance program consistent with sound business practice.

 

13.6                         In making the determination referenced in Paragraph 13.5, PHS shall take into account the normal course of such commercial development programs conducted with sound and reasonable business practices and judgment and the annual reports submitted by Licensee under Paragraph 9.2. Prior to invoking termination or modification of this Agreement under Paragraph 13.5, PHS shall give written notice to Licensee providing Licensee specific notice of, and a ninety (90) day opportunity to respond to. PHS’ concerns as to the items referenced in 13.5(a)-13.5(g). If Licensee fails to alleviate PHS’ concerns as to the items referenced in 13.5(a)-13.5(g) or fails to initiate corrective action to PHS’ satisfaction, PHS may terminate this Agreement.

 

13.7                         PHS reserves the right according to 35 U.S.C. §209(d)(3) to terminate or modify this Agreement if it is determined that the action is necessary to meet the requirements for public use specified by federal regulations issued after the date of the license and these requirements are not reasonably satisfied by Licensee.

 

13.8                         Within thirty (30) days of receipt of written notice of PHS’ unilateral decision to modify or terminate this Agreement, Licensee may, consistent with the provisions of 37 CFR §404.11, appeal the decision by written submission to the designated PHS official. The decision of the designated PHS official shall be the final agency decision. Licensee may thereafter exercise any and all administrative or judicial remedies that maybe available.

 

B- 4



 

EXHIBIT C

 

Approved Activities

 

1.      Estimated Product expenses for January to April 2013:

 

 

 

Subtotal

 

Approved Activity

 

Jan to Feb

 

Mar to Apr

 

TOTAL
Jan to Apr

 

Assay Development

 

 

 

 

 

€207K

 

Seeds & product tests

 

€25K

 

€177K

 

 

 

PoC in HB mice

 

 

 

€5K

 

 

 

 

 

 

 

 

 

 

 

Note: these are not the full costs

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Non clinical

 

 

 

 

 

€430K

 

NHP GLP tox/ safety

 

€40K

 

€40K

 

 

 

Mouse GLP tox/ biodistribution

 

€150K

 

€150K

 

 

 

Germline transmission

 

€50K

 

 

 

 

 

 

 

 

 

 

 

 

 

Note: these are not the full costs

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Manufacturing

 

 

 

 

 

€69K

 

Phase I material procurement

 

€46

 

€23K

 

 

 

 

 

 

 

 

 

 

 

Note: these are not the full costs

 

 

 

 

 

 

 

 

TOTAL Product Development Costs (out-of-pocket) for January to April 2013 = €706K

 

C- 1



 

EXHIBIT D

 

Certain Terms for HemB Supply and Distribution Agreement

 

Transfer Price : Chiesi shall purchase the Product for the greater of (a) forty percent (40%) of the Average Net Sales Price (in Brazil and Mexico)or, as the case may be, thirty percent (30%) of the Average Net Sales Price (in all other countries of the Territory) (Note: definition needs to conform with uniQure’s in-licenses) or (b) uniQure’s Fully Loaded Cost of Goods plus twenty percent (20%) mark up, for each patient dose sold (the “Product Transfer Price”). The Product Manufacturing Cost Reimbursement associated with each patient dose of the Product sold by Chiesi shall be credited against the Product Transfer Price.  The “Product Manufacturing Cost Reimbursement” for each patient dose of the Product shall be uniQure’s Fully Loaded Costs of Goods.

 

Commercial Supply : Prior to initiation of the Pivotal Study, the Parties shall negotiate in good faith the HemB Supply and Distribution Agreement for commercial material with the financial terms described above and other terms expected to be substantially similar to the Commercialization Agreement. For the avoidance of doubt, the provisions set forth in Section 2.6 of the Commercialization Agreement shall be included in the HemB Supply and Distribution Agreement.

 

“Fully Loaded Costs of Goods” shall be determined according to the principles applied in the Commercialization Agreement, in particular Schedule 2.3 thereof.

 

Indemnification :

 

Indemnification by Chiesi.  Chiesi shall indemnify, defend and hold harmless uniQure and its Affiliates, and its and their respective directors, officers, employees and agents, from and against any and all Losses, arising out of or resulting from any and all Third Party Claims to the extent based upon:

 

(a)           any breach of any representation, warranty or covenant made by, or any material obligation of, Chiesi under the HemB Supply and Distribution Agreement;

 

(b)           the gross negligence, recklessness or willful misconduct of Chiesi or its Affiliates and its or their respective directors, officers, employees and agents; or

 

(c)           any theory of product liability (including without limitation tort, warranty, or strict liability) that is applicable in the Territory with respect to the death, personal injury, or illness of any Person in the Territory, and arising directly from Chiesi’s or its Affiliates’ or Sub-distributors’ Commercialization of the Product in the Territory;

 

provided that Chiesi shall not be obligated to indemnify uniQureto the extent uniQure is required to indemnify Chiesi under the HemB Supply and Distribution Agreement.

 

Indemnification by uniQure.  uniQure shall indemnify, defend and hold harmless Chiesi and its Affiliates, and its and their respective directors, officers, employees and agents, from and against any and all Losses, arising out of or resulting from any and all Claims to the extent based upon:

 

D- 1



 

(a)           any breach of any representation, warranty or covenant made by, or any material obligation of, uniQure under the HemB Supply and Distribution Agreement;

 

(b)           the gross negligence, recklessness or willful misconduct of uniQure or its Affiliates and its or their respective directors, officers, employees and agents;

 

(c)           any theory of product liability (including without limitation tort, warranty, or strict liability) that is applicable in the Territory with respect to the death, personal injury, or illness of any Person in the Territory, and arising directly from uniQure’s or its Affiliates’ design, Manufacture, storage, release and handling of the Product; or

 

(d)           Claims that the (i) Commercialization of the Product; or (ii) exercise of any rights or licenses granted to Chiesi and its Affiliates in accordance with the HemB Supply and Distribution Agreement; violates or infringes upon the Intellectual Property Rights of any Third Party;

 

provided that uniQure shall not be obligated to indemnify Chiesi to the extent Chiesi is required to indemnify uniQure under the HemB Supply and Distribution Agreement.

 

Trademark : The Product will be Commercialized by Chiesi in the Field in the Territory exclusively under a trademark mutually agreed between the Parties. Such trademark shall be owned by uniQure and shall be licensed to Chiesi for Commercialization of the Product in the Field in the Territory. Section 2.2(a) of the Commercialization Agreement shall apply mutatis mutandis .

 

D- 2



 

EXHIBIT E

 

Licensed Patents

 

UniQure patent portfolio — AMT-060 (Hemophilia B)

 

UniQure Ref.

 

Country

 

Application No.

 

Publication No.

 

Case Status

 

Filing Date

 

Owner

Baculoviral vectors comprising repeated coding sequences with differential codon biases

AMT-P107

 

Europe

 

07113257.5

 

 

 

Withdraw Before Publication

 

26-Jul-2007

 

Amsterdam Molecular Therapeutics B.V.

AMT-P107

 

PCT

 

PCT/NL2008/050512

 

 

 

Abandoned

 

25-Jul-2008

 

Amsterdam Molecular Therapeutics B.V.

AMT-P107

 

Brazile

 

PI 0814459-1

 

 

 

Examination Requested

 

25-Jul-2008

 

Amsterdam Molecular Therapeutics B.V.

AMT-P107

 

Eurasian Patent Organization

 

201070184

 

 

 

Pending

 

25-Jul-2008

 

Amsterdam Molecular Therapeutics B.V.

AMT-P107

 

Europe

 

08779058.0

 

2173888

 

Application filed

 

25-Jul-2008

 

UniQure IP B.V.

AMT-P107

 

MX

 

MX/a/2010/000944

 

 

 

Published

 

25-Jul-2008

 

Amsterdam Molecular Therapeutics (AMT) B.V.

 

 

 

 

 

 

 

 

 

 

 

 

 

Removal of contaminating viruses from AAV preparations

AMT-P113

 

EP

 

11180594.1

 

 

 

Abandoned

 

08-Sep-2011

 

UniQure IP B.V.

AMT-P113

 

PCT

 

PCT/NL2012/050619

 

 

 

Pending

 

07-Sep-2012

 

UniQure IP B.V.

AMT-P113

 

WO

 

 

 

WO/2013/036118

 

Published

 

14-Mrt-2013

 

UniQure IP B.V.

 

In-Licensed by uniQure AMT-060 (Hemophilia B)

 

Application No.

 

Publication No.

 

Case
Status

 

Filing Date

 

Registration
Date

 

Registration No.

 

Priority Details Date

 

Owners

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

PRODUCTION OF ADENO-ASSOCIATED VIRUS IN INSECT CELLS

PHS, Bethesda, USA

 

09/986,618

 

 

 

 

 

09.Nov.2001

 

 

 

 

 

09.Nov.2001

 

 

L-107-2007/0

 

02795604.4

 

EP1572893

 

granted

 

08.Nov.2002

 

07.Jan.2009

 

EP1572893

 

09.Nov.2001

 

 

 

 

 

 

 

 

 

 

 

 

 

 

DE, FR, GB

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

SPODOPTERA FRUGIPERDA SINGLE CELL SUSPENSION CELL LINE IN SERUM-FREE MEDIA, METHODS OF PRODUCING AND USING THE SAME

PSC, Meriden, USA

 

99950093.7

 

EP1119612

 

granted

 

4 oct 1999

 

21.Jul.2010

 

EP1119612

 

8-oct-1998

 

 

 

 

 

 

 

 

 

 

 

 

 

 

AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, IE, IT, U, LU, MC, NL, PT

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

AAVS VECTOR AND USES THEREOF

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

PHS, Bethesda, USA

 

99926036.7

 

EP1082413

 

Granted

 

28 may 1999

 

23.Jul.2008

 

EP1082413

 

28 may 1998

 

 

L-116-2011/0

 

 

 

 

 

 

 

 

 

 

 

DE, ES, FR, GB, IT, NL

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

IMPROVED EXPRESSION OF FACTOR FIX IN GENE THERAPY VECTORS

St. Jude Children’s Research Hospital

 

05796011.4

 

EP1804839

 

Granted

 

09.09.205

 

14.03.2012

 

EP1804839

 

22 Sep 04

 

 

Memphis, USA

 

 

 

 

 

 

 

 

 

 

 

AT, BE, CH, DE, DK, ES, FR, GB, IT, LI, LU, NL, SE, TR

SJ-04-0024A

 

12150318.9

 

Ep2438931

 

Pending

 

04.01.2012

 

 

 

EP2438931

 

22.Sep 04

 

 

 

E- 1



 

Note:

 

Patents with the Case Status “Abandoned” or “Withdrawn” are listed for information purposes but are not part of the Licensed Patents.

 

E- 2



 

EXHIBIT F

 

uniQure FTE Limits

 

Unless otherwise agreed in writing as part of the then current Development Plan and Budget, the number of uniQure FTEs will be no higher than:

 

·                   25.4 FTEs in 2013

·                   20.2 FTEs in 2014

·                   25 FTEs in 2015

·                   25 FTEs in 2016

·                   10 FTEs in 2017

 

Unless otherwise agreed in writing as part of the then current Development Plan and Budget, the number of Chiesi FTEs will be no higher than:

 

·                   6.8 FTEs in 2013

·                   5.8 FTEs in 2014

·                   8 FTEs in 2015

·                   8 FTEs in 2016

·                   4 FTEs in 2017

 

F- 1



 

EXHIBIT G

 

Technology Transfer

 

The following is a non-exhaustive list describing key steps which the Parties would typically envisage for a transfer of the Manufacturing of the Product to another manufacturing site during Product Development:

 

Steps

 

Estimated Timelines

·                   if transferred to a Third Party manufacturer: select and contract manufacturer party

 

4 months

·                   process transfer (on paper)

 

2 months

·                   obtain time slot

 

2 months

·                   process validation

 

7 months

Total

 

15 months

 

G- 1



 

EXHIBIT H

 

Disclosure Schedule to Section 11.2(e)

 

uniQure has become aware of a potential license grant from PHS to Sangamo BioSciences, Inc., Richmond, USA. uniQure understands that such license may potentially conflict with the scope of the exclusive license granted from PHS to uniQure under the PHS 2011 Agreement. uniQure is currently in the process of further investigating this potential issue and shall timely update Chiesi on the outcome of its inquiry and any further steps taken, if necessary.

 

G- 1


Exhibit 31.1

 

Certification of Chief Executive Officer

 

I, Matthew Kapusta, certify that:

 

1.             I have reviewed this Quarterly Report on Form 10-Q of uniQure N.V.;

 

2.             Based on my knowledge, this report does not contain any untrue statement of a material fact or omit to state a material fact necessary to make the statements made, in light of the circumstances under which such statements were made, not misleading with respect to the period covered by this report;

 

3.             Based on my knowledge, the financial statements, and other financial information included in this report, fairly present in all material respects the financial condition, results of operations and cash flows of the registrant as of, and for, the periods presented in this report;

 

4.             The registrant’s other certifying officer and I are responsible for establishing and maintaining disclosure controls and procedures (as defined in Exchange Act Rules 13a-15(e) and 15d-15(e)) and internal controls over financial reporting (as defined in Exchange Act Rules 13a-15(f) and 15d-15(f)) for the registrant and have:

 

(a)           Designed such disclosure controls and procedures, or caused such disclosure controls and procedures to be designed under our supervision, to ensure that material information relating to the registrant, including its consolidated subsidiaries, is made known to us by others within those entities, particularly during the period in which this report is being prepared;

 

(b)           Designed such internal control over financial reporting, or caused such internal control over financial reporting to be designed under our supervision, to provide reasonable assurance regarding the reliability of financial reporting and the preparation of financial statements for external purposes in accordance with generally accepted accounting principles;

 

(c)           Evaluated the effectiveness of the registrant’s disclosure controls and procedures and presented in this report our conclusions about the effectiveness of the disclosure controls and procedures, as of the end of the period covered by this report based on such evaluation; and

 

(d)           Disclosed in this report any change in the registrant’s internal control over financial reporting that occurred during the registrant’s most recent fiscal quarter (the registrant’s fourth fiscal quarter in the case of an annual report) that has materially affected, or is reasonably likely to materially affect, the registrant’s internal control over financial reporting; and

 

5.             The registrant’s other certifying officer and I have disclosed, based on our most recent evaluation of internal control over financial reporting, to the registrant’s auditors and the audit committee of the registrant’s board of directors (or persons performing the equivalent functions):

 

(a)           All significant deficiencies and material weaknesses in the design or operation of internal control over financial reporting which are reasonably likely to adversely affect the registrant’s ability to record, process, summarize and report financial information; and

 

(b)           Any fraud, whether or not material, that involves management or other employees who have a significant role in the registrant’s internal control over financial reporting.

 

 

 

By:

/s/ MATTHEW KAPUSTA

 

 

 

 

 

Matthew Kapusta

 

 

Chief Executive Officer

 

 

May 9, 2017

 


Exhibit 31.2

 

Certification of Chief Financial Officer

 

I, Matthew Kapusta, certify that:

 

1.             I have reviewed this Quarterly Report on Form 10-Q of uniQure N.V.;

 

2.             Based on my knowledge, this report does not contain any untrue statement of a material fact or omit to state a material fact necessary to make the statements made, in light of the circumstances under which such statements were made, not misleading with respect to the period covered by this report;

 

3.             Based on my knowledge, the financial statements, and other financial information included in this report, fairly present in all material respects the financial condition, results of operations and cash flows of the registrant as of, and for, the periods presented in this report;

 

4.             The registrant’s other certifying officer and I are responsible for establishing and maintaining disclosure controls and procedures (as defined in Exchange Act Rules 13a-15(e) and 15d-15(e)) and internal controls over financial reporting (as defined in Exchange Act Rules 13a-15(f) and 15d-15(f)) for the registrant and have:

 

(a)           Designed such disclosure controls and procedures, or caused such disclosure controls and procedures to be designed under our supervision, to ensure that material information relating to the registrant, including its consolidated subsidiaries, is made known to us by others within those entities, particularly during the period in which this report is being prepared;

 

(b)           Designed such internal control over financial reporting, or caused such internal control over financial reporting to be designed under our supervision, to provide reasonable assurance regarding the reliability of financial reporting and the preparation of financial statements for external purposes in accordance with generally accepted accounting principles;

 

(c)           Evaluated the effectiveness of the registrant’s disclosure controls and procedures and presented in this report our conclusions about the effectiveness of the disclosure controls and procedures, as of the end of the period covered by this report based on such evaluation; and

 

(d)           Disclosed in this report any change in the registrant’s internal control over financial reporting that occurred during the registrant’s most recent fiscal quarter (the registrant’s fourth fiscal quarter in the case of an annual report) that has materially affected, or is reasonably likely to materially affect, the registrant’s internal control over financial reporting; and

 

5.             The registrant’s other certifying officer and I have disclosed, based on our most recent evaluation of internal control over financial reporting, to the registrant’s auditors and the audit committee of the registrant’s board of directors (or persons performing the equivalent functions):

 

(a)           All significant deficiencies and material weaknesses in the design or operation of internal control over financial reporting which are reasonably likely to adversely affect the registrant’s ability to record, process, summarize and report financial information; and

 

(b)           Any fraud, whether or not material, that involves management or other employees who have a significant role in the registrant’s internal control over financial reporting.

 

 

 

By:

/s/ MATTHEW KAPUSTA

 

 

 

 

 

Matthew Kapusta

 

 

Chief Financial Officer

 

 

May 9, 2017

 


Exhibit 32.1

 

CERTIFICATION PURSUANT TO 18 U.S.C. SECTION 1350

AS ADOPTED PURSUANT TO SECTION 906 OF THE SARBANES-OXLEY ACT OF 2002

 

In connection with this Quarterly Report of uniQure N.V. (the “Company”) on Form 10-Q for the period ended March 31, 2017, as filed with the Securities and Exchange Commission on the date hereof (the “Report”), the undersigned, Matthew Kapusta, Chief Executive Officer and Chief Financial Officer of the Company, hereby certifies, pursuant to 18 U.S.C. Section 1350, as adopted pursuant to Section 906 of the Sarbanes-Oxley Act of 2002, that:

 

1              the Report fully complies with the requirements of Section 13(a) or 15(d) of the Securities Exchange Act of 1934; and

 

2              the information contained in the Report fairly presents, in all material respects, the financial condition and results of operations of the Company.

 

 

By:

/s/ MATTHEW KAPUSTA

 

 

 

 

 

Matthew Kapusta

 

 

Chief Executive Officer and

 

 

Chief Financial Officer

 

 

May 9, 2017

 

A signed original of this written statement required by Section 906 has been provided to uniQure N.V. and will be retained by uniQure N.V. and furnished to the SEC or its staff upon request.