10-Q
0001631574one yearQ2falseone year--12-31five years0001631574wve:AtTheMarketEquityProgramMember2022-01-012022-06-300001631574wve:TakedaPharmaceuticalCompanyLimitedMemberwve:CollaborationAndSharePurchaseAgreementsMember2018-04-012018-04-300001631574us-gaap:CommonStockMember2022-01-012022-03-310001631574us-gaap:RetainedEarningsMember2021-04-012021-06-300001631574us-gaap:SeriesAPreferredStockMember2021-03-310001631574us-gaap:ShortTermInvestmentsMemberus-gaap:FairValueMeasurementsRecurringMember2022-06-300001631574us-gaap:CommonStockMemberwve:PerformanceBasedRestrictedStockUnitsMember2022-04-012022-04-300001631574us-gaap:AccumulatedOtherComprehensiveIncomeMember2021-06-300001631574us-gaap:FairValueInputsLevel1Memberwve:RestrictedCashMemberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001631574us-gaap:FairValueInputsLevel2Memberus-gaap:ShortTermInvestmentsMember2022-06-300001631574us-gaap:CommonStockMember2021-03-310001631574us-gaap:EmployeeStockOptionMember2021-01-012021-06-300001631574us-gaap:AccumulatedOtherComprehensiveIncomeMember2021-03-310001631574wve:CollaborationAndLicenseAgreementMemberwve:TakedaPharmaceuticalCompanyLimitedMember2018-04-012018-04-0200016315742022-08-030001631574us-gaap:AccumulatedOtherComprehensiveIncomeMember2022-03-310001631574us-gaap:FairValueInputsLevel2Memberus-gaap:ShortTermInvestmentsMemberus-gaap:FairValueMeasurementsRecurringMember2022-06-300001631574us-gaap:RetainedEarningsMember2022-03-310001631574wve:CollaborationAndLicenseAgreementMemberwve:TakedaPharmaceuticalCompanyLimitedMember2018-04-012018-04-3000016315742021-01-012021-06-300001631574wve:PreFundedWarrantsMember2022-06-3000016315742022-01-012022-06-300001631574wve:CollaborationAndLicenseAgreementMemberwve:CategoryTwoProgramsMemberwve:TakedaPharmaceuticalCompanyLimitedMember2021-10-152021-10-150001631574wve:TakedaPharmaceuticalCompanyLimitedMemberwve:CollaborationAndSharePurchaseAgreementsMember2018-04-300001631574us-gaap:RetainedEarningsMember2022-06-300001631574wve:CategoryOneProgramsMemberwve:CollaborationAndLicenseAgreementMemberwve:TakedaPharmaceuticalCompanyLimitedMember2021-04-012021-06-300001631574wve:TwoThousandTwentyOneEquityIncentivePlanMember2022-06-300001631574us-gaap:RetainedEarningsMember2021-12-310001631574us-gaap:AdditionalPaidInCapitalMember2022-03-310001631574wve:CambridgeMassachusettsMember2022-01-012022-06-300001631574wve:CategoryOneProgramsMemberwve:CollaborationAndLicenseAgreementMemberwve:TakedaPharmaceuticalCompanyLimitedMember2022-06-300001631574wve:CollaborationAndLicenseAgreementMemberwve:CategoryTwoProgramsMemberwve:TakedaPharmaceuticalCompanyLimitedMember2018-04-012018-04-300001631574wve:TwoThousandFourteenEquityIncentivePlanMembersrt:MaximumMemberus-gaap:RestrictedStockUnitsRSUMember2022-01-012022-06-300001631574us-gaap:AccumulatedOtherComprehensiveIncomeMember2021-12-310001631574us-gaap:SeriesAPreferredStockMember2022-01-012022-06-3000016315742021-12-3100016315742021-01-012021-03-310001631574wve:PreFundedWarrantsMember2022-06-162022-06-160001631574wve:TakedaPharmaceuticalCompanyLimitedMember2018-04-012018-04-300001631574wve:CollaborationAndLicenseAgreementMembersrt:MinimumMemberwve:TakedaPharmaceuticalCompanyLimitedMember2018-02-012018-02-280001631574us-gaap:SeriesAPreferredStockMember2021-01-012021-06-300001631574us-gaap:RetainedEarningsMember2022-04-012022-06-300001631574us-gaap:AdditionalPaidInCapitalMember2022-01-012022-03-3100016315742022-06-300001631574wve:CategoryOneProgramsMemberwve:CollaborationAndLicenseAgreementMemberwve:TakedaPharmaceuticalCompanyLimitedMember2021-10-152021-10-150001631574wve:ScientificAdvisorMemberwve:ConsultingAgreementMember2022-01-012022-06-3000016315742022-01-012022-03-310001631574wve:AtTheMarketEquityProgramMember2021-04-012021-06-300001631574wve:PerformanceBasedRestrictedStockUnitsMember2022-04-012022-04-300001631574us-gaap:RestrictedStockUnitsRSUMember2021-01-012021-06-300001631574wve:CambridgeMassachusettsMember2022-06-300001631574wve:PerformanceBasedRestrictedStockUnitsMember2022-06-300001631574us-gaap:EmployeeStockOptionMember2022-01-012022-06-300001631574us-gaap:AccumulatedOtherComprehensiveIncomeMember2022-06-300001631574wve:TwoThousandNineteenEmployeeSharePurchasePlanMember2022-01-012022-06-300001631574us-gaap:AccumulatedOtherComprehensiveIncomeMember2022-01-012022-03-310001631574wve:RestrictedCashMemberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001631574us-gaap:SeriesAPreferredStockMember2020-12-310001631574wve:TwoThousandFourteenEquityIncentivePlanMembersrt:MaximumMember2022-01-012022-06-300001631574wve:PreFundedWarrantsMember2022-06-160001631574wve:TwoThousandFourteenEquityIncentivePlanMembersrt:MinimumMemberus-gaap:RestrictedStockUnitsRSUMember2022-01-012022-06-300001631574us-gaap:FairValueMeasurementsRecurringMember2021-12-310001631574wve:RestrictedCashMemberus-gaap:FairValueMeasurementsRecurringMember2022-06-300001631574us-gaap:RetainedEarningsMember2020-12-3100016315742022-03-310001631574wve:CategoryOneProgramsMemberwve:CollaborationAndLicenseAgreementMemberwve:TakedaPharmaceuticalCompanyLimitedMember2021-01-012021-06-300001631574wve:AtTheMarketEquityProgramMember2021-01-012021-03-3100016315742021-03-310001631574us-gaap:CommonStockMember2021-12-310001631574us-gaap:SeriesAPreferredStockMember2021-06-3000016315742021-10-010001631574us-gaap:EmployeeStockOptionMember2022-01-012022-06-300001631574us-gaap:AdditionalPaidInCapitalMember2021-06-300001631574us-gaap:AdditionalPaidInCapitalMember2020-12-310001631574wve:LexingtonMassachusettsMember2016-09-260001631574us-gaap:CommonStockMember2022-03-310001631574us-gaap:CommonStockMember2022-04-012022-06-300001631574us-gaap:AccumulatedOtherComprehensiveIncomeMember2020-12-3100016315742022-04-012022-06-3000016315742020-12-310001631574wve:PreFundedWarrantsMembersrt:MaximumMember2022-06-160001631574wve:CambridgeMassachusettsMember2015-04-300001631574us-gaap:CommonStockMember2021-04-012021-06-300001631574us-gaap:RetainedEarningsMember2021-06-300001631574wve:LexingtonMassachusettsMember2021-12-310001631574wve:AtTheMarketEquityProgramMember2022-01-012022-03-310001631574wve:June2022OfferingMember2022-06-162022-06-160001631574us-gaap:AdditionalPaidInCapitalMember2022-06-300001631574wve:June2022OfferingMember2022-06-160001631574wve:LexingtonMassachusettsMember2022-06-300001631574us-gaap:CommonStockMemberwve:AtTheMarketEquityProgramMember2021-01-012021-03-310001631574wve:LexingtonMassachusettsMember2022-01-012022-06-300001631574us-gaap:CommonStockMemberwve:AtTheMarketEquityProgramMember2021-04-012021-06-300001631574us-gaap:AdditionalPaidInCapitalMember2021-01-012021-03-310001631574us-gaap:RetainedEarningsMember2021-03-310001631574wve:TimeBasedRestrictedStockUnitsMember2022-01-012022-06-300001631574us-gaap:SeriesAPreferredStockMember2022-06-300001631574us-gaap:FairValueInputsLevel1Memberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001631574us-gaap:CommonStockMember2020-12-310001631574us-gaap:FairValueMeasurementsRecurringMember2022-06-300001631574us-gaap:AdditionalPaidInCapitalMember2022-04-012022-06-3000016315742021-04-012021-06-300001631574wve:CategoryOneProgramsMemberwve:CollaborationAndLicenseAgreementMemberwve:TakedaPharmaceuticalCompanyLimitedMember2022-04-012022-06-300001631574us-gaap:CommonStockMember2021-06-300001631574us-gaap:FairValueInputsLevel2Memberus-gaap:ShortTermInvestmentsMember2021-12-310001631574wve:CambridgeMassachusettsMember2021-12-310001631574us-gaap:AdditionalPaidInCapitalMember2021-03-310001631574us-gaap:AccumulatedOtherComprehensiveIncomeMember2022-04-012022-06-300001631574us-gaap:CommonStockMember2022-06-300001631574wve:TwoThousandFourteenEquityIncentivePlanMembersrt:MinimumMember2022-01-012022-06-300001631574wve:CategoryOneProgramsMemberwve:CollaborationAndLicenseAgreementMemberwve:TakedaPharmaceuticalCompanyLimitedMember2022-01-012022-06-300001631574wve:CategoryOneProgramsMemberwve:CollaborationAndLicenseAgreementMemberwve:TakedaPharmaceuticalCompanyLimitedMember2018-02-012022-03-310001631574us-gaap:FairValueInputsLevel1Memberwve:RestrictedCashMemberus-gaap:FairValueMeasurementsRecurringMember2022-06-300001631574us-gaap:CommonStockMemberwve:AtTheMarketEquityProgramMember2022-01-012022-03-310001631574us-gaap:RetainedEarningsMember2021-01-012021-03-310001631574us-gaap:SeriesAPreferredStockMember2021-12-310001631574us-gaap:AdditionalPaidInCapitalMember2021-12-310001631574wve:AtTheMarketEquityProgramMember2021-01-012021-06-300001631574wve:TwoThousandNineteenEmployeeSharePurchasePlanMember2022-06-300001631574us-gaap:RestrictedStockUnitsRSUMember2022-01-012022-06-300001631574wve:PreFundedWarrantsMembersrt:MinimumMember2022-06-162022-06-160001631574us-gaap:CommonStockMember2021-01-012021-03-310001631574wve:CollaborationAndLicenseAgreementMemberwve:CategoryTwoProgramsMemberwve:TakedaPharmaceuticalCompanyLimitedMember2022-01-012022-06-300001631574us-gaap:FairValueInputsLevel2Memberus-gaap:FairValueMeasurementsRecurringMember2022-06-300001631574us-gaap:AdditionalPaidInCapitalMember2021-04-012021-06-300001631574us-gaap:FairValueInputsLevel1Memberus-gaap:FairValueMeasurementsRecurringMember2022-06-300001631574wve:CategoryOneProgramsMemberwve:CollaborationAndLicenseAgreementMember2018-04-012018-04-300001631574us-gaap:RetainedEarningsMember2022-01-012022-03-310001631574wve:CategoryOneProgramsMemberwve:CollaborationAndLicenseAgreementMemberwve:TakedaPharmaceuticalCompanyLimitedMember2018-04-012018-04-3000016315742021-06-300001631574us-gaap:SeriesAPreferredStockMember2022-03-310001631574us-gaap:AccumulatedOtherComprehensiveIncomeMember2021-01-012021-03-310001631574wve:CategoryOneProgramsMemberwve:CollaborationAndLicenseAgreementMemberwve:TakedaPharmaceuticalCompanyLimitedMember2021-12-31iso4217:USDxbrli:sharesxbrli:purewve:Targetxbrli:sharesiso4217:USD

 

 

UNITED STATES

SECURITIES AND EXCHANGE COMMISSION

Washington, D.C. 20549

 

FORM 10-Q

 

(Mark One)

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

For the quarterly period ended June 30, 2022

     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: 001-37627

 

WAVE LIFE SCIENCES LTD.

(Exact name of registrant as specified in its charter)

 

Singapore

(State or other jurisdiction of incorporation or organization)

 

00-0000000

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

 

 

 

7 Straits View #12-00, Marina One East Tower

Singapore

(Address of principal executive offices)

 

018936

(Zip Code)

 

+65 6236 3388

(Registrant’s telephone number, including area code)

 

Securities registered pursuant to Section 12(b) of the Act:

 

Title of each class

Trading symbol

Name of each exchange on which registered

$0 Par Value Ordinary Shares

WVE

The Nasdaq Global Market

 

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 ☒ No ☐

 

Indicate by check mark whether the registrant has submitted electronically every Interactive Data File required to be submitted 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 such files). Yes ☒ No ☐

 

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

 

Large accelerated filer

 

Accelerated filer

Non-accelerated filer

 

Smaller reporting company

 

 

 

Emerging growth company

 

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. ☐

 

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

 

The number of outstanding ordinary shares of the registrant as of August 3, 2022 was 86,820,111
.

 

 


 

WAVE LIFE SCIENCES LTD.

QUARTERLY REPORT ON FORM 10-Q

TABLE OF CONTENTS

 

 

 

Page

PART I - FINANCIAL INFORMATION

 

5

Item 1. Financial Statements

 

5

Unaudited Consolidated Balance Sheets

 

5

Unaudited Consolidated Statements of Operations and Comprehensive Loss

 

6

Unaudited Consolidated Statements of Series A Preferred Shares and Shareholders' Equity (Deficit)

 

7

Unaudited Consolidated Statements of Cash Flows

 

9

Notes to Unaudited Consolidated Financial Statements

 

10

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

 

20

Item 3. Quantitative and Qualitative Disclosures About Market Risk

 

33

Item 4. Controls and Procedures

 

33

PART II - OTHER INFORMATION

 

34

Item 1. Legal Proceedings

 

34

Item 1A. Risk Factors

 

34

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

 

34

Item 3. Defaults Upon Senior Securities

 

34

Item 4. Mine Safety Disclosures

 

34

Item 5. Other Information

 

34

Item 6. Exhibits

 

35

 

2


 

As used in this Quarterly Report on Form 10-Q, unless otherwise stated or the context otherwise indicates, references to “Wave,” the “Company,” “we,” “our,” “us” or similar terms refer to Wave Life Sciences Ltd. and our wholly-owned subsidiaries.

Special Note Regarding Forward-Looking Statements

This Quarterly Report on Form 10-Q contains forward-looking statements within the meaning of Section 27A of the Securities Act of 1933, as amended (the “Securities Act”), and Section 21E of the Securities Exchange Act of 1934, as amended (the “Exchange Act”), that relate to future events or to our future operations or financial performance. Any forward-looking statement involves known and unknown risks, uncertainties and other factors that may cause our actual results, levels of activity, performance or achievements to differ materially from any future results, levels of activity, performance or achievements expressed or implied by such forward-looking statement. In some cases, forward-looking statements are identified by the words “anticipate,” “believe,” “continue,” “could,” “estimate,” “expect,” “future,” “goals,” “intend,” “likely,” “may,” “might,” “ongoing,” “objective,” “plan,” “potential,” “predict,” “project,” “seek,” “should,” “strategy,” “target,” “will” and “would” or the negative of these terms, or other comparable terminology intended to identify statements about the future, although not all forward-looking statements contain these identifying words. Forward-looking statements include statements, other than statements of historical fact, about, among other things: our ability to fund our future operations; our financial position, revenues, costs, expenses, uses of cash and capital requirements; our need for additional financing or the period for which our existing cash resources will be sufficient to meet our operating requirements; the success, progress, number, scope, cost, duration, timing or results of our research and development activities, preclinical studies and clinical trials, including the timing for initiation or completion of or availability of results from any preclinical studies and clinical trials or for submission, review or approval of any regulatory filing; the timing of, and our ability to, obtain and maintain regulatory approvals for any of our product candidates; the potential benefits that may be derived from any of our product candidates; our strategies, prospects, plans, goals, expectations, forecasts or objectives; the success of our collaborations with third parties; any payment that our collaboration partners may make to us; our ability to identify and develop new product candidates; our intellectual property position; our commercialization, marketing and manufacturing capabilities and strategy; our ability to develop sales and marketing capabilities; our ability to identify, recruit and retain key personnel; our financial performance; developments and projections relating to our competitors in the industry; our liquidity and working capital requirements; the expected impact of new accounting standards; and our expectations regarding the impact of COVID-19 and variants thereof on our business, including on our research and development activities, preclinical studies and clinical trials, supply of drug product, and workforce.

Although we believe that we have a reasonable basis for each forward-looking statement contained in this report, we caution you that these statements are based on our estimates or projections of the future that are subject to known and unknown risks and uncertainties and other important factors that may cause our actual results, level of activity, performance or achievements expressed or implied by any forward-looking statement to differ. These risks, uncertainties and other factors include, among other things, our critical accounting policies; the ability of our preclinical studies to produce data sufficient to support the filing of global clinical trial applications and the timing thereof; our ability to continue to build and maintain the company infrastructure and personnel needed to achieve our goals; the clinical results and timing of our programs, which may not support further development of our product candidates; actions of regulatory agencies, which may affect the initiation, timing and progress of clinical trials; our effectiveness in managing current and future clinical trials and regulatory processes; the success of our platform in identifying viable candidates; the continued development and acceptance of nucleic acid therapeutics as a class of drugs; our ability to demonstrate the therapeutic benefits of our stereopure candidates in clinical trials, including our ability to develop candidates across multiple therapeutic modalities; our ability to obtain, maintain and protect intellectual property; our ability to enforce our patents against infringers and defend our patent portfolio against challenges from third parties; our ability to fund our operations and to raise additional capital as needed; competition from others developing therapies for similar uses; the severity and duration of the COVID-19 pandemic; the COVID-19 pandemic, and variants thereof, may negatively impact the conduct of, and the timing of enrollment, completion and reporting with respect to, our clinical trials; any other impacts on our business as a result of or related to the COVID-19 pandemic, as well as other risks and uncertainties under the caption “Risk Factors” contained in this Quarterly Report on Form 10-Q and in other filings we make with the Securities and Exchange Commission.

 

3


 

Each forward-looking statement contained in this report is based on a combination of facts and factors currently known by us and our expectations of the future, about which we cannot be certain. As a result of these factors, we cannot assure you that the forward-looking statements in this report will prove to be accurate. Furthermore, if our forward-looking statements prove to be inaccurate, the inaccuracy may be material. In light of the significant uncertainties in these forward-looking statements, these statements should not be regarded as representations or warranties by us or any other person that we will achieve our objectives and plans in any specified timeframe, or at all. We caution you not to place undue reliance on any forward-looking statement.

In addition, any forward-looking statement in this report represents our views only as of the date of this report and should not be relied upon as representing our views as of any subsequent date. We anticipate that subsequent events and developments may cause our views to change. Although we may elect to update these forward-looking statements publicly at some point in the future, we undertake no obligation to publicly update any forward-looking statements, whether as a result of new information, future events or otherwise, except as required by applicable law. Our forward-looking statements do not reflect the potential impact of any future acquisitions, mergers, dispositions, joint ventures or investments we may make.

 

The Wave Life Sciences Ltd. and Wave Life Sciences Pte. Ltd. names, the Wave Life Sciences mark, PRISM and the other registered and pending trademarks, trade names and service marks of Wave Life Sciences Ltd. appearing in this Form 10-Q are the property of Wave Life Sciences Ltd. This Form 10-Q also contains additional trade names, trademarks and service marks belonging to Wave Life Sciences Ltd. and to other companies. We do not intend our use or display of other parties’ trademarks, trade names or service marks to imply, and such use or display should not be construed to imply, a relationship with, or endorsement or sponsorship of us by, these other parties. Solely for convenience, the trademarks and trade names in this Form 10-Q are referred to without the ® and ™ symbols, but such reference should not be construed as any indicator that their respective owners will not assert, to the fullest extent under applicable law, their rights thereto.

4


 

PART I - FINANCIAL INFORMATION

Item 1. Financial Statements

WAVE LIFE SCIENCES LTD.

UNAUDITED CONSOLIDATED BALANCE SHEETS

(In thousands, except share amounts)

 

 

 

June 30, 2022

 

 

December 31, 2021

 

Assets

 

 

 

 

 

 

Current assets:

 

 

 

 

 

 

Cash and cash equivalents

 

$

123,163

 

 

$

150,564

 

Short-term investments

 

 

25,000

 

 

 

 

Prepaid expenses

 

 

5,396

 

 

 

6,584

 

Other current assets

 

 

6,555

 

 

 

5,416

 

Total current assets

 

 

160,114

 

 

 

162,564

 

Long-term assets:

 

 

 

 

 

 

Property and equipment, net

 

 

19,808

 

 

 

22,266

 

Operating lease right-of-use assets

 

 

28,791

 

 

 

18,378

 

Restricted cash

 

 

3,651

 

 

 

3,651

 

Other assets

 

 

955

 

 

 

148

 

Total long-term assets

 

 

53,205

 

 

 

44,443

 

Total assets

 

$

213,319

 

 

$

207,007

 

Liabilities, Series A preferred shares and shareholders’ equity

 

 

 

 

 

 

Current liabilities:

 

 

 

 

 

 

Accounts payable

 

$

11,168

 

 

$

7,281

 

Accrued expenses and other current liabilities

 

 

11,085

 

 

 

14,861

 

Current portion of deferred revenue

 

 

37,466

 

 

 

37,098

 

Current portion of operating lease liability

 

 

4,363

 

 

 

4,961

 

Total current liabilities

 

 

64,082

 

 

 

64,201

 

Long-term liabilities:

 

 

 

 

 

 

Deferred revenue, net of current portion

 

 

75,256

 

 

 

77,479

 

Operating lease liability, net of current portion

 

 

35,160

 

 

 

24,955

 

Total long-term liabilities

 

 

110,416

 

 

 

102,434

 

Total liabilities

 

$

174,498

 

 

$

166,635

 

Series A preferred shares, no par value; 3,901,348 shares
   issued and outstanding at June 30, 2022 and December 31, 2021

 

$

7,874

 

 

$

7,874

 

Shareholders’ equity:

 

 

 

 

 

 

Ordinary shares, no par value; 86,724,658 and 59,841,116 shares
   issued and outstanding at June 30, 2022 and December 31, 2021, respectively

 

$

802,449

 

 

$

749,851

 

Additional paid-in capital

 

 

113,169

 

 

 

87,980

 

Accumulated other comprehensive income (loss)

 

 

(47

)

 

 

181

 

Accumulated deficit

 

 

(884,624

)

 

 

(805,514

)

Total shareholders’ equity

 

$

30,947

 

 

$

32,498

 

Total liabilities, Series A preferred shares and shareholders’ equity

 

$

213,319

 

 

$

207,007

 

 

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

 

5


 

WAVE LIFE SCIENCES LTD.

UNAUDITED CONSOLIDATED STATEMENTS OF OPERATIONS AND COMPREHENSIVE LOSS

(In thousands, except share and per share amounts)

 

 

 

Three Months Ended June 30,

 

 

Six Months Ended June 30,

 

 

 

2022

 

 

2021

 

 

2022

 

 

2021

 

Revenue

 

$

375

 

 

$

2,776

 

 

$

2,125

 

 

$

2,776

 

Operating expenses:

 

 

 

 

 

 

 

 

 

 

 

 

Research and development

 

 

29,733

 

 

 

31,635

 

 

 

57,203

 

 

 

65,028

 

General and administrative

 

 

12,806

 

 

 

10,969

 

 

 

25,180

 

 

 

21,047

 

Total operating expenses

 

 

42,539

 

 

 

42,604

 

 

 

82,383

 

 

 

86,075

 

Loss from operations

 

 

(42,164

)

 

 

(39,828

)

 

 

(80,258

)

 

 

(83,299

)

Other income, net:

 

 

 

 

 

 

 

 

 

 

 

 

Dividend income and interest income, net

 

 

124

 

 

 

8

 

 

 

150

 

 

 

19

 

Other income, net

 

 

744

 

 

 

1,054

 

 

 

998

 

 

 

2,050

 

Total other income, net

 

 

868

 

 

 

1,062

 

 

 

1,148

 

 

 

2,069

 

Loss before income taxes

 

 

(41,296

)

 

 

(38,766

)

 

 

(79,110

)

 

 

(81,230

)

Income tax provision

 

 

 

 

 

 

 

 

 

 

 

 

Net loss

 

$

(41,296

)

 

$

(38,766

)

 

$

(79,110

)

 

$

(81,230

)

Net loss per share attributable to ordinary
   shareholders—basic and diluted

 

$

(0.62

)

 

$

(0.78

)

 

$

(1.25

)

 

$

(1.65

)

Weighted-average ordinary shares used in
   computing net loss per share attributable to
   ordinary shareholders—basic and diluted

 

 

66,479,293

 

 

 

49,973,185

 

 

 

63,514,426

 

 

 

49,220,140

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Other comprehensive loss:

 

 

 

 

 

 

 

 

 

 

 

 

Net loss

 

$

(41,296

)

 

$

(38,766

)

 

$

(79,110

)

 

$

(81,230

)

Foreign currency translation

 

 

(142

)

 

 

 

 

 

(228

)

 

 

(120

)

Comprehensive loss

 

$

(41,438

)

 

$

(38,766

)

 

$

(79,338

)

 

$

(81,350

)

 

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

 

6


 

WAVE LIFE SCIENCES LTD.

UNAUDITED CONSOLIDATED STATEMENTS OF SERIES A PREFERRED SHARES AND SHAREHOLDERS’ EQUITY (DEFICIT)

 

(In thousands, except share amounts)

 

 

 

Series A
Preferred Shares

 

 

 

Ordinary Shares

 

 

Additional
Paid-In-

 

 

Accumulated
Other
Comprehensive

 

 

Accumulated

 

 

Total
Shareholders’

 

 

 

Shares

 

 

Amount

 

 

 

Shares

 

 

Amount

 

 

Capital

 

 

Income (Loss)

 

 

Deficit

 

 

Equity

 

Balance at December 31, 2020

 

 

3,901,348

 

 

$

7,874

 

 

 

 

48,778,678

 

 

$

694,085

 

 

$

71,573

 

 

$

389

 

 

$

(683,269

)

 

$

82,778

 

Issuance of ordinary shares
   pursuant to the at-the-market
   equity program, net

 

 

 

 

 

 

 

 

 

844,796

 

 

 

8,028

 

 

 

 

 

 

 

 

 

 

 

 

8,028

 

Share-based compensation

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

4,063

 

 

 

 

 

 

 

 

 

4,063

 

Vesting of RSUs

 

 

 

 

 

 

 

 

 

155,184

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Option exercises

 

 

 

 

 

 

 

 

 

31,957

 

 

 

200

 

 

 

 

 

 

 

 

 

 

 

 

200

 

Issuance of ordinary shares
   under the ESPP

 

 

 

 

 

 

 

 

 

44,036

 

 

 

336

 

 

 

 

 

 

 

 

 

 

 

 

336

 

Other comprehensive loss

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

(120

)

 

 

 

 

 

(120

)

Net loss

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

(42,464

)

 

 

(42,464

)

Balance at March 31, 2021

 

 

3,901,348

 

 

$

7,874

 

 

 

 

49,854,651

 

 

$

702,649

 

 

$

75,636

 

 

$

269

 

 

$

(725,733

)

 

$

52,821

 

Issuance of ordinary shares
   pursuant to the at-the-market
   equity program, net

 

 

 

 

 

 

 

 

 

718,179

 

 

 

5,065

 

 

 

 

 

 

 

 

 

 

 

 

5,065

 

Share-based compensation

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

2,722

 

 

 

 

 

 

 

 

 

2,722

 

Vesting of RSUs

 

 

 

 

 

 

 

 

 

3,636

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Net loss

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

(38,766

)

 

 

(38,766

)

Balance at June 30, 2021

 

 

3,901,348

 

 

$

7,874

 

 

 

 

50,576,466

 

 

$

707,714

 

 

$

78,358

 

 

$

269

 

 

$

(764,499

)

 

$

21,842

 

 

 

7


 

WAVE LIFE SCIENCES LTD.

UNAUDITED CONSOLIDATED STATEMENTS OF SERIES A PREFERRED SHARES AND SHAREHOLDERS’ EQUITY (DEFICIT) CONTINUED

 

(In thousands, except share amounts)

 

 

 

Series A
Preferred Shares

 

 

 

Ordinary Shares

 

 

Additional
Paid-In-

 

 

Accumulated
Other
Comprehensive

 

 

Accumulated

 

 

Total
Shareholders’

 

 

 

Shares

 

 

Amount

 

 

 

Shares

 

 

Amount

 

 

Capital

 

 

Income (Loss)

 

 

Deficit

 

 

Equity (Deficit)

 

Balance at December 31, 2021

 

 

3,901,348

 

 

$

7,874

 

 

 

 

59,841,116

 

 

$

749,851

 

 

$

87,980

 

 

$

181

 

 

$

(805,514

)

 

$

32,498

 

Issuance of ordinary shares
   pursuant to the at-the-market
   equity program, net

 

 

 

 

 

 

 

 

 

458,092

 

 

 

1,167

 

 

 

 

 

 

 

 

 

 

 

 

1,167

 

Share-based compensation

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

3,971

 

 

 

 

 

 

 

 

 

3,971

 

Vesting of RSUs

 

 

 

 

 

 

 

 

 

468,226

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Option exercises

 

 

 

 

 

 

 

 

 

15,000

 

 

 

37

 

 

 

 

 

 

 

 

 

 

 

 

37

 

Issuance of ordinary shares
   under the ESPP

 

 

 

 

 

 

 

 

 

77,534

 

 

 

174

 

 

 

 

 

 

 

 

 

 

 

 

174

 

Other comprehensive loss

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

(86

)

 

 

 

 

 

(86

)

Net loss

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

(37,814

)

 

 

(37,814

)

Balance at March 31, 2022

 

 

3,901,348

 

 

$

7,874

 

 

 

 

60,859,968

 

 

$

751,229

 

 

$

91,951

 

 

$

95

 

 

$

(843,328

)

 

$

(53

)

Issuance of ordinary shares,
   net of offering costs

 

 

 

 

 

 

 

 

 

25,464,483

 

 

 

51,220

 

 

 

 

 

 

 

 

 

 

 

 

51,220

 

Issuance of pre-funded warrants,
   net of offering costs

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

14,268

 

 

 

 

 

 

 

 

 

14,268

 

Share-based compensation

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

6,950

 

 

 

 

 

 

 

 

 

6,950

 

Vesting of RSUs

 

 

 

 

 

 

 

 

 

400,207

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Other comprehensive loss

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

(142

)

 

 

 

 

 

(142

)

Net loss

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

(41,296

)

 

 

(41,296

)

Balance at June 30, 2022

 

 

3,901,348

 

 

$

7,874

 

 

 

 

86,724,658

 

 

$

802,449

 

 

$

113,169

 

 

$

(47

)

 

$

(884,624

)

 

$

30,947

 

 

The accompanying notes are an integral part of the consolidated financial statements.

8


 

WAVE LIFE SCIENCES LTD.

UNAUDITED CONSOLIDATED STATEMENTS OF CASH FLOWS

(In thousands)

 

 

 

Six Months Ended June 30,

 

 

 

2022

 

 

2021

 

Cash flows from operating activities

 

 

 

 

 

 

Net loss

 

$

(79,110

)

 

$

(81,230

)

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

 

Amortization of right-of-use assets

 

 

1,593

 

 

 

1,043

 

Depreciation of property and equipment

 

 

3,425

 

 

 

3,857

 

Share-based compensation expense

 

 

10,921

 

 

 

6,785

 

Changes in operating assets and liabilities:

 

 

 

 

 

 

Accounts receivable

 

 

 

 

 

30,000

 

Prepaid expenses

 

 

1,188

 

 

 

1,246

 

Other assets

 

 

(1,946

)

 

 

(3,475

)

Accounts payable

 

 

3,264

 

 

 

(5,251

)

Accrued expenses and other current liabilities

 

 

(3,776

)

 

 

(2,048

)

Deferred revenue

 

 

(1,855

)

 

 

(2,776

)

Operating lease liabilities

 

 

(2,399

)

 

 

(1,792

)

Other non-current liabilities

 

 

 

 

 

(135

)

Net cash used in operating activities

 

 

(68,695

)

 

 

(53,776

)

Cash flows from investing activities

 

 

 

 

 

 

Purchases of property and equipment

 

 

(700

)

 

 

(447

)

Proceeds from the sale of property and equipment

 

 

106

 

 

 

 

Purchase of short-term investments

 

 

(50,000

)

 

 

 

Proceeds from the maturity of short-term investments

 

 

25,000

 

 

 

 

Net cash used in investing activities

 

 

(25,594

)

 

 

(447

)

Cash flows from financing activities

 

 

 

 

 

 

Proceeds from issuance of ordinary shares, net of offering costs

 

 

51,464

 

 

 

 

Proceeds from issuance pre-funded warrants, net of offering costs

 

 

14,336

 

 

 

 

Proceeds from issuance of ordinary shares pursuant to the
   at-the-market equity program, net of offering costs

 

 

1,105

 

 

 

13,150

 

Proceeds from the exercise of share options

 

 

37

 

 

 

200

 

Proceeds from the ESPP

 

 

174

 

 

 

336

 

Net cash provided by financing activities

 

 

67,116

 

 

 

13,686

 

Effect of foreign exchange rates on cash, cash equivalents and restricted cash

 

 

(228

)

 

 

(120

)

Net decrease in cash, cash equivalents and restricted cash

 

 

(27,401

)

 

 

(40,657

)

Cash, cash equivalents and restricted cash, beginning of period

 

 

154,215

 

 

 

188,148

 

Cash, cash equivalents and restricted cash, end of period

 

$

126,814

 

 

$

147,491

 

Supplemental disclosure of cash flow information:

 

 

 

 

 

 

Increase in operating lease right-of-use assets and
   lease liabilities related to lease extension

 

$

12,006

 

 

$

 

Offering costs in accounts payable at period end

 

$

311

 

 

$

57

 

 

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

 

9


 

Wave Life Sciences Ltd.

Notes to Unaudited Consolidated Financial Statements

1. THE COMPANY

Organization

Wave Life Sciences Ltd. (together with its subsidiaries, “Wave” or the “Company”) is a clinical-stage genetic medicines company committed to delivering life-changing treatments for people battling devastating diseases. PRISM, Wave’s proprietary discovery and drug development platform, enables the precise design, optimization and production of novel stereopure oligonucleotides. Wave has built a genetic toolkit comprised of multiple therapeutic modalities, including RNase-H mediated silencing, RNAi, splicing, and RNA base editing, all of which leverage learnings and optimizations from the PRISM platform and allow Wave to design built-for-purpose molecules to optimally address disease biology.

The Company was incorporated in Singapore on July 23, 2012 and has its principal U.S. office in Cambridge, Massachusetts. The Company was incorporated with the purpose of combining two commonly held companies, Wave Life Sciences USA, Inc. (“Wave USA”), a Delaware corporation (formerly Ontorii, Inc.), and Wave Life Sciences Japan, Inc. (“Wave Japan”), a company organized under the laws of Japan (formerly Chiralgen., Ltd.), which occurred on September 13, 2012. On May 31, 2016, Wave Life Sciences Ireland Limited (“Wave Ireland”) was formed as a wholly-owned subsidiary of Wave Life Sciences Ltd. On April 3, 2017, Wave Life Sciences UK Limited (“Wave UK”) was formed as a wholly-owned subsidiary of Wave Life Sciences Ltd.

The Company’s primary activities since inception have been developing and evolving PRISM to design, develop and commercialize oligonucleotide therapeutics, advancing the Company’s differentiated neurology portfolio, as well as exploring other therapeutic areas of interest, building the Company’s research, development and manufacturing capabilities, advancing programs into the clinic, furthering clinical development of such clinical-stage programs, building the Company’s intellectual property, and assuring adequate capital to support these activities.

Liquidity

Since its inception, the Company has not generated any product revenue and has incurred recurring net losses. To date, the Company has primarily funded its operations through private placements of debt and equity securities, public and other registered offerings of its equity securities and collaborations with third parties. Until the Company can generate significant revenue from product sales, if ever, the Company expects to continue to finance operations through a combination of public or private equity or debt financings or other sources, which may include collaborations with third parties. Adequate additional financing may not be available to the Company on acceptable terms, or at all. The inability to raise capital as and when needed would have a negative impact on the Company’s financial condition and ability to pursue its business strategy.

As of June 30, 2022, the Company had cash, cash equivalents and short-term investments of $148.2 million. The Company expects that its existing cash, cash equivalents and short-term investments will be sufficient to fund its operations for at least the next twelve months. The Company has based this expectation on assumptions that may prove to be incorrect, and the Company may use its available capital resources sooner than it currently expects. If the Company’s anticipated operating results are not achieved in future periods, planned expenditures may need to be further reduced in order to extend the time period over which the then-available resources would be able to fund the Company’s operations. In addition, the Company may elect to raise additional funds before it needs them if the conditions for raising capital are favorable due to market conditions or strategic considerations, even if the Company expects it has sufficient funds for its current or future operating plans.

Risks and Uncertainties

The Company is subject to risks common to companies in the biotechnology industry including, but not limited to, new technological innovations, protection of proprietary technology, maintaining internal manufacturing capabilities, dependence on key personnel, compliance with government regulations and the need to obtain additional financing. The Company’s therapeutic programs will require significant additional research and development efforts, including extensive preclinical and clinical testing and regulatory approval, prior to commercialization of any product candidates. These efforts require significant amounts of additional capital, adequate personnel infrastructure and extensive compliance-reporting capabilities. There can be no assurance that the Company’s research and development efforts will be successful, that adequate protection for the Company’s intellectual property will be obtained, that any products developed will obtain necessary government regulatory approval or that any approved products will be commercially viable. Even if the Company’s product development efforts are successful, it is uncertain when, if ever, the Company will generate significant revenue from product sales. The Company operates in an environment of rapid change in technology and substantial competition from pharmaceutical and biotechnology companies.

10


 

Basis of Presentation

The Company has prepared the accompanying consolidated financial statements in conformity with generally accepted accounting principles in the United States (“U.S. GAAP”) and in U.S. dollars.

2. SIGNIFICANT ACCOUNTING POLICIES

The significant accounting policies described in the Company’s audited financial statements as of and for the year ended December 31, 2021, and the notes thereto, which are included in the Company’s Annual Report on Form 10-K for the year ended December 31, 2021, filed with the Securities and Exchange Commission (“SEC”) on March 3, 2022, as amended (the “2021 Annual Report on Form 10-K”), have had no material changes during the six months ended June 30, 2022, except as described below.

Fair Value of Financial Instruments

The Company is required to disclose information on all assets and liabilities reported at fair value that enables an assessment of the inputs used in determining the reported fair values. The fair value hierarchy is a hierarchy of inputs used in measuring fair value that maximizes the use of observable inputs and minimizes the use of unobservable inputs by requiring that the observable inputs be used when available. Observable inputs are inputs that market participants would use in pricing the financial instrument based on market data obtained from sources independent of the Company. Unobservable inputs are inputs that reflect the Company’s assumptions about the inputs that market participants would use in pricing the financial instrument and are developed based on the information available in the circumstances. The fair value hierarchy applies only to the valuation inputs used in determining the reported fair value of the investments and is not a measure of the investment credit quality. The hierarchy defines three levels of valuation inputs:

Level 1—Unadjusted quoted prices in active markets that are accessible at the measurement date of identical, unrestricted assets.

Level 2—Quoted prices for similar assets, or inputs that are observable, either directly or indirectly, for substantially the full term through corroboration with observable market data. Level 2 includes investments valued at quoted prices adjusted for legal or contractual restrictions specific to the security.

Level 3—Pricing inputs are unobservable for the asset, that is, inputs that reflect the reporting entity’s own assumptions about the assumptions market participants would use in pricing the asset. Level 3 includes private investments that are supported by little or no market activity.

Cash and Cash Equivalents

The Company considers all highly liquid securities with original final maturities of three months or less from the date of purchase to be cash equivalents. Cash and cash equivalents are comprised of funds in cash and money market accounts.

Short-Term Investments

The Company considers all time deposits with original maturities of more than three months from the date of purchase to be short-term investments.

Concentration of Credit Risk

Cash, cash equivalents, restricted cash and short-term investments are financial instruments that potentially subject the Company to concentration of credit risk. The Company uses several financial institutions to maintain its cash, cash equivalents, restricted cash and short-term investments, all of which are high quality, accredited financial institutions and, accordingly, such funds are subject to minimal credit risk. The Company has not experienced any losses in such accounts and management believes that the Company is not exposed to significant credit risk due to the financial position of the depository institutions in which those deposits are held. The Company has no financial instruments with off-balance sheet risk of loss.

11


 

Unaudited Interim Financial Data

The accompanying interim consolidated balance sheet as of June 30, 2022, the related interim consolidated statements of operations and comprehensive loss for the three and six months ended June 30, 2022 and 2021, the consolidated statements of Series A preferred shares and shareholders’ equity (deficit) for the three months ended March 31, and June 30, 2022 and 2021, the consolidated statements of cash flows for the six months ended June 30, 2022 and 2021, and the related interim information contained within the notes to the unaudited consolidated financial statements have been prepared in accordance with the rules and regulations of the SEC for interim financial information. Accordingly, they do not include all of the information and the notes required by U.S. GAAP for complete financial statements. The financial data and other information disclosed in these notes related to the three and six months ended June 30, 2022 and 2021 are unaudited. In the opinion of management, the unaudited interim consolidated financial statements reflect all adjustments, consisting of normal and recurring adjustments, necessary for the fair presentation of the Company’s financial position and results of operations for the three and six months ended June 30, 2022 and 2021. The results of operations for the interim periods are not necessarily indicative of the results to be expected for the year ending December 31, 2022 or any other interim period or future year or period.

3. FAIR VALUE MEASUREMENTS

The following table sets forth the Company’s financial assets that are measured at fair value on a recurring basis:

 

 

 

Total

 

 

Level 1

 

 

Level 2

 

 

Level 3

 

 

 

(in thousands)

 

June 30, 2022

 

 

 

 

 

 

 

 

 

 

 

 

Cash and cash equivalents

 

$

123,163

 

 

$

123,163

 

 

$

 

 

$

 

Short-term investments

 

 

25,000

 

 

 

 

 

 

25,000

 

 

 

 

Restricted cash

 

 

3,651

 

 

 

3,651

 

 

 

 

 

 

 

Total

 

$

151,814

 

 

$

126,814

 

 

$

25,000

 

 

$

 

December 31, 2021

 

 

 

 

 

 

 

 

 

 

 

 

Cash and cash equivalents

 

$

150,564

 

 

$

150,564

 

 

$

 

 

$

 

Short-term investments

 

 

 

 

 

 

 

 

 

 

 

 

Restricted cash

 

 

3,651

 

 

 

3,651

 

 

 

 

 

 

 

Total

 

$

154,215

 

 

$

154,215

 

 

$

 

 

$

 

 

There have been no transfers between fair value levels during the three months ended June 30, 2022.

Cash, cash equivalents and restricted cash are Level 1 assets which are comprised of funds held in checking and money market accounts. Short-term investments are Level 2 assets which are comprised of time deposits with original maturities of more than three months. The Company determined that the fair value of its short-term investments is $25.0 million as of June 30, 2022, which approximates the carrying value of the term deposits. There were no short-term investments as of December 31, 2021, as the term deposits that constitute the Company’s short-term investments were purchased during the six months ended June 30, 2022. The carrying amounts of accounts payable and accrued expenses approximate their fair values due to their short-term maturities.

12


 

4. SHARE-BASED COMPENSATION

The Wave Life Sciences Ltd. 2021 Equity Incentive Plan (the “2021 Plan”) was approved by the Company’s shareholders and went into effect on August 10, 2021. The 2021 Plan serves as the successor to the Wave Life Sciences Ltd. 2014 Equity Incentive Plan, as amended (the “2014 Plan”), such that outstanding awards granted under the 2014 Plan continue to be governed by the terms of the 2014 Plan, but no awards may be made under the 2014 Plan after August 10, 2021. The aggregate number of ordinary shares authorized for issuance of awards under the 2021 Plan is 5,450,000 ordinary shares, plus the number of ordinary shares underlying any awards under the 2014 Plan that are forfeited, cancelled or otherwise terminated (other than by exercise or withheld by the Company to satisfy any tax withholding obligation) on or after August 10, 2021.

The 2021 Plan authorizes (and the 2014 Plan previously authorized) the Company’s board of directors or a committee of the board of directors to, among other things, grant non-qualified share options, restricted awards, which include restricted shares and restricted share units (“RSUs”), and performance awards to eligible employees and directors of the Company.

Options generally vest over periods of one to four years, and any options that are forfeited or cancelled are available to be granted again. The contractual life of options is generally five or ten years from the grant date. RSUs can be time-based or performance-based. Time-based RSUs generally vest over a period of one to four years. The vesting of performance-based RSUs is contingent on the achievement of certain performance milestones. Any RSUs that are forfeited are available to be granted again.

In March 2021, the compensation committee of the Company’s board of directors approved an amendment and restatement of the Company’s outstanding 2019 performance-based RSUs to add an additional milestone to the existing milestones. In 2021, the Company also granted performance-based RSUs with the same terms to certain employees who did not receive the 2019 performance-based RSUs. This modification did not result in any incremental expense.

In April 2022, the Company determined that a performance-based RSU milestone was achieved and consequently 50% of the outstanding performance-based RSUs vested, which resulted in the issuance of 384,646 ordinary shares. During the three months ended June 30, 2022, the Company recorded share-based compensation expense of approximately $3.8 million, which represents all of the expense related to the achievement of this performance-based RSU milestone. The Company did not recognize any expense related to the other performance-based RSU milestones, as the remaining milestones were not considered probable of achievement as of June 30, 2022.

During the six months ended June 30, 2022, the Company granted 2,037,175 options and 55,075 time-based RSUs to employees.

As of June 30, 2022, 1,561,172 ordinary shares remained available for future grant under the 2021 Plan.

The Wave Life Sciences Ltd. 2019 Employee Share Purchase Plan (“ESPP”) allows all full-time and certain part-time employees to purchase the Company’s ordinary shares at a discount to fair market value. Eligible employees may enroll in a six-month offering period beginning on or about January 15th and July 15th every year. Shares are purchased at a price equal to 85% of the lower of the fair market value of the Company’s ordinary shares on the first business day or the last business day of an offering period. During the six months ended June 30, 2022, 77,534 ordinary shares were issued under the ESPP. As of June 30, 2022, there were 804,940 ordinary shares available for issuance under the ESPP.

13


 

5. COLLABORATION AGREEMENTS

Takeda Collaboration and Equity Agreements

In February 2018, Wave USA and Wave UK entered into a global strategic collaboration (the “Takeda Collaboration”) with Takeda Pharmaceutical Company Limited (“Takeda”), pursuant to which Wave USA, Wave UK and Takeda agreed to collaborate on the research, development and commercialization of oligonucleotide therapeutics for disorders of the Central Nervous System (“CNS”). The Takeda Collaboration provides the Company with at least $230.0 million in committed cash and Takeda with the option to co-develop and co-commercialize the Company’s CNS development programs in (1) Huntington’s disease (“HD”); (2) amyotrophic lateral sclerosis (“ALS”) and frontotemporal dementia (“FTD”); and (3) the Company’s discovery-stage program targeting ATXN3 for the treatment of spinocerebellar ataxia 3 (“SCA3”) (collectively, “Category 1 Programs”). In addition, the Takeda Collaboration provided Takeda the right to exclusively license multiple preclinical programs for CNS disorders, including Alzheimer’s disease and Parkinson’s disease (collectively, “Category 2 Programs”). In April 2018, the Takeda Collaboration became effective and Takeda paid the Company $110.0 million as an upfront payment. Takeda also agreed to fund the Company’s research and preclinical activities in the amount of $60.0 million during the four-year research term and to reimburse the Company for any collaboration-budgeted research and preclinical expenses incurred by the Company that exceed that amount.

Simultaneously with Wave USA and Wave UK’s entry into the collaboration and license agreement with Takeda (the “Takeda Collaboration Agreement”), the Company entered into a share purchase agreement with Takeda (the “Takeda Equity Agreement,” and together with the Takeda Collaboration Agreement, the “Takeda Agreements”) pursuant to which it agreed to sell to Takeda 1,096,892 of its ordinary shares at a purchase price of $54.70 per share. In April 2018, the Company closed the Takeda Equity Agreement and received aggregate cash proceeds of $60.0 million. The Company did not incur any material costs in connection with the issuance of shares.

With respect to Category 1 Programs, the Company will be responsible for researching and developing products and companion diagnostics for Category 1 Programs through completion of the first proof of mechanism study for such products. Takeda will have an exclusive option for each target and all associated products and companion diagnostics for such target, which it may exercise at any time through completion of the proof of mechanism study. If Takeda exercises this option, the Company will receive an opt-in payment and will lead manufacturing and joint clinical co-development activities and Takeda will lead joint co-commercial activities in the United States and all commercial activities outside of the United States. Global costs and potential profits will be shared 50:50 and the Company will be eligible to receive development and commercial milestone payments. In addition to its 50% profit share, the Company is eligible to receive option exercise fees and development and commercial milestone payments for each of the Category 1 Programs.

With respect to Category 2 Programs, the Company granted Takeda the right to exclusively license multiple preclinical programs during a four-year research term (subject to limited extension for programs that were initiated prior to the expiration of the research term, in accordance with the Takeda Collaboration Agreement) (“Category 2 Research Term”). During that term, the Takeda Collaboration provided that the parties may collaborate on preclinical programs for up to six targets at any one time. The Company was responsible for researching and preclinically developing products and companion diagnostics directed to the agreed upon targets through completion of Investigational New Drug application (“IND”)-enabling studies in the first major market country. Thereafter, Takeda would have an exclusive worldwide license to develop and commercialize products and companion diagnostics directed to such targets, subject to the Company’s retained rights to lead manufacturing activities for products directed to such targets. Takeda agreed to fund the Company’s research and preclinical activities in the amount of $60.0 million during the research term and reimburse the Company for any collaboration-budgeted research and preclinical expenses incurred by the Company that exceeded that amount. The Company was also eligible to receive tiered high single-digit to mid-teen royalties on Takeda’s global commercial sales of products from each Category 2 Program.

Under the Takeda Collaboration Agreement, each party granted to the other party specific intellectual property licenses to enable the other party to perform its obligations and exercise its rights under the Takeda Collaboration Agreement, including license grants to enable each party to conduct research, development and commercialization activities pursuant to the terms of the Takeda Collaboration Agreement.

The term of the Takeda Collaboration Agreement commenced on April 2, 2018 and, unless terminated earlier, will continue until the date on which: (i) with respect to each Category 1 Program target for which Takeda does not exercise its option, the expiration or termination of the development program with respect to such target; (ii) with respect to each Category 1 Program target for which Takeda exercises its option, the date on which neither party is researching, developing or manufacturing any products or companion diagnostics directed to such target; or (iii) with respect to each Category 2 Program target, the date on which royalties are no longer payable with respect to products directed to such target.

14


 

Takeda may terminate the Takeda Collaboration Agreement for convenience on 180 days’ notice, in its entirety or on a target-by-target basis. Subject to certain exceptions, each party has the right to terminate the Takeda Collaboration Agreement on a target-by-target basis if the other party, or a third party related to such party, challenges the patentability, enforceability or validity of any patents within the licensed technology that cover any product or companion diagnostic that is subject to the Takeda Collaboration Agreement. In the event of any material breach of the Takeda Collaboration Agreement by a party, subject to cure rights, the other party may terminate the Takeda Collaboration Agreement in its entirety if the breach relates to all targets or on a target-by-target basis if the breach relates to a specific target. In the event that Takeda and its affiliates cease development, manufacturing and commercialization activities with respect to compounds or products subject to the Takeda Collaboration Agreement and directed to a particular target, the Company may terminate the Takeda Collaboration Agreement with respect to such target. Either party may terminate the Takeda Collaboration Agreement for the other party’s insolvency. In certain termination circumstances, the Company would receive a license from Takeda to continue researching, developing and manufacturing certain products, and companion diagnostics.

The Takeda Collaboration is managed by a joint steering committee (“JSC”) in which both parties are represented equally. The JSC is tasked with overseeing the scientific progression of each Category 1 Program and, prior to the Amendment (discussed below), the Category 2 Programs.

The Company assessed this arrangement in accordance with ASC Topic 606, Revenue from Contracts with Customers (“ASC 606”) and concluded that the contract counterparty, Takeda, is a customer for Category 1 Programs prior to Takeda exercising its option, and for Category 2 Programs during the Category 2 Research Term. The Company identified the following material promises under the arrangement: (1) the non-exclusive, royalty-free research and development license for each Category 1 Program; (2) the research and development services for each Category 1 Program through completion of the first proof of mechanism study; (3) the exclusive option to license, co-develop and co-commercialize each Category 1 Program; (4) the right to exclusively license the Category 2 Programs; and (5) the research and preclinical development services of the Category 2 Programs through completion of IND-enabling studies. The research and development services for each Category 1 Program were determined to not be distinct from the research and development license and should therefore be combined into a single performance obligation for each Category 1 Program. The research and preclinical development services for the Category 2 Programs were determined to not be distinct from the exclusive licenses for the Category 2 Programs and should therefore be combined into a single performance obligation.

Additionally, the Company determined that the exclusive option for each Category 1 Program was priced at a discount, and, as such, provide material rights to Takeda, representing three separate performance obligations. Based on these assessments, the Company identified seven performance obligations in the Takeda Collaboration Agreement: (1) research and development services through completion of the first proof of mechanism and non-exclusive research and development license for HD; (2) research and development services through completion of the first proof of mechanism and non-exclusive research and development license for ALS and FTD; (3) research and development services through completion of the first proof of mechanism and non-exclusive research and development license for SCA3; (4) the material right provided for the exclusive option to license, co-develop and co-commercialize HD; (5) the material right provided for the exclusive option to license, co-develop and co-commercialize ALS and FTD; (6) the material right provided for the exclusive option to license, co-develop and co-commercialize SCA3; and (7) the research and preclinical development services and right to exclusively license the Category 2 Programs.

At the outset of the arrangement, the transaction price included the $110.0 million upfront consideration received and the $60.0 million of committed research and preclinical funding for the Category 2 Programs. The Company determined that the Takeda Collaboration Agreement did not contain a significant financing component. The option exercise fees to license, co-develop and co-commercialize each Category 1 Program that may be received are excluded from the transaction price until each customer option is exercised. The potential milestone payments were excluded from the transaction price, as all milestone amounts were fully constrained at the inception of the Takeda Collaboration Agreement. The Company will reevaluate the transaction price at the end of each reporting period and as uncertain events are resolved or other changes in circumstances occur, if necessary, will adjust its estimate of the transaction price.

15


 

The Company allocated the transaction price to the performance obligations on a relative standalone selling price basis. For the performance obligations associated with the research and development services through completion of the first proof of mechanism and non-exclusive research and development license for HD; the research and development services through completion of the first proof of mechanism and non-exclusive research and development license for ALS and FTD; the research and development services through completion of the first proof of mechanism and non-exclusive research and development license for SCA3; and the research and preclinical development services and right to exclusively license the Category 2 Programs, the Company determined the standalone selling price using estimates of the costs to perform the research and development services, including expected internal and external costs for services and supplies, adjusted to reflect a profit margin. The total estimated cost of the research and development services reflected the nature of the services to be performed and the Company’s best estimate of the length of time required to perform the services. For the performance obligations associated with the material right provided for the exclusive option to license, co-develop and co-commercialize HD; the material right provided for the exclusive option to license, co-develop and co-commercialize ALS and FTD; and the material right provided for the exclusive option to license, co-develop and co-commercialize SCA3, the Company estimated the standalone fair value of the option to license each Category 1 Program utilizing an adjusted market assessment approach, and determined that any standalone fair value in excess of the amounts to be paid by Takeda associated with each option represented a material right.

Revenue associated with the research and development services for each Category 1 Program performance obligation is being recognized as the research and development services are provided using an input method, according to the costs incurred on each Category 1 Program and the total costs expected to be incurred to satisfy each Category 1 Program performance obligation. Revenue associated with the research and preclinical development services for the Category 2 Programs performance obligation is being recognized as the research and preclinical development services are provided using an input method, according to the costs incurred on Category 2 Programs and the total costs expected to be incurred to satisfy the performance obligation. The transfer of control for these performance obligations occurs over time and, in management’s judgment, this input method is the best measure of progress towards satisfying the performance obligations. The amount allocated to the material right for each Category 1 Program option will be recognized on the date that Takeda exercises each respective option, or immediately as each option expires unexercised. The amounts received that have not yet been recognized as revenue are recorded in deferred revenue on the Company’s consolidated balance sheet.

On October 15, 2021, Wave USA, Wave UK and Takeda entered into the Second Amendment to the Takeda Collaboration Agreement (the “Amendment”), which discontinued the Category 2 component of the Takeda Collaboration. Pursuant to the Amendment, Takeda agreed to pay the Company an additional $22.5 million as full payment for reimbursable Category 2 Program collaboration-budgeted research and preclinical expenses. The Category 1 Programs under the Takeda Collaboration Agreement remain in effect and are unchanged by the Amendment.

Through June 30, 2022, the Company had recognized revenue of approximately $79.8 million as collaboration revenue under the Takeda Collaboration Agreement. During the three and six months ended June 30, 2022, the Company recognized revenue of $0.3 million and $1.9 million under the Takeda Collaboration Agreement, respectively. During the three and six months ended June 30, 2021, the Company recognized revenue of approximately $2.8 million under the Takeda Collaboration Agreement.

The aggregate amount of the transaction price allocated to the Company’s unsatisfied and partially unsatisfied performance obligations and recorded in deferred revenue as of December 31, 2021 was $114.2 million, of which $37.1 million was included in current liabilities. The aggregate amount of the transaction price allocated to the Company’s unsatisfied and partially unsatisfied performance obligations and recorded in deferred revenue at June 30, 2022 is $112.7 million, of which $37.5 million is included in current liabilities. The Company expects to recognize revenue for the portion of the deferred revenue that relates to the research and development services for each Category 1 Program as costs are incurred, over the remaining research term. The Company expects to recognize revenue for the portion of the deferred revenue that relates to the material right for each Category 1 Program option upon Takeda’s exercise of such option, or immediately as each option expires unexercised.

16


 

6. SHAREHOLDERS’ EQUITY

June 2022 Offering

On June 16, 2022, the Company closed an underwritten offering (the “June 2022 Offering”) in which the Company issued and sold 25,464,483 of the Company’s ordinary shares at a price of $2.15 per share and pre-funded warrants (the “Pre-Funded Warrants”) to purchase up to 7,093,656 of the Company’s ordinary shares at an offering price of $2.1499 per Pre-Funded Warrant, which represents the per share offering price for the ordinary shares less the $0.0001 per share exercise price for each Pre-Funded Warrant. These Pre-Funded Warrants were recorded as a component of shareholders’ equity within additional paid-in capital. The gross proceeds to the Company were $70.0 million before deducting underwriting discounts and commissions and other offering expenses. The net proceeds of the June 2022 Offering were approximately $65.5 million.

The Pre-Funded Warrants are exercisable at any time after their original issuance and on or prior to the five-year anniversary of the original issuance date. A holder of Pre-Funded Warrants may not exercise the warrant if the holder, together with its affiliates, would beneficially own more than 19.99% of the number of ordinary shares outstanding or more than 19.99% of the combined voting power of the Company’s securities outstanding immediately after giving effect to such exercise, unless and until shareholder approval is obtained.

7. NET LOSS PER ORDINARY SHARE

The Company applies the two-class method to calculate its basic and diluted net loss per share attributable to ordinary shareholders, as its Series A preferred shares are participating securities. The two-class method is an earnings allocation formula that treats a participating security as having rights to earnings that otherwise would have been available to ordinary shareholders.

As of June 30, 2022, there are 7,093,656 vested and exercisable Pre-Funded Warrants outstanding to purchase ordinary shares for the exercise price of $0.0001 per share. The Pre-Funded Warrants are included in the weighted-average shares outstanding used in the calculation of basic net loss per share as the exercise price is negligible and the warrants are fully vested and exercisable.

Basic loss per share is computed by dividing net loss attributable to ordinary shareholders and Pre-Funded Warrant holders by the weighted-average number of ordinary shares and Pre-Funded Warrants outstanding.

The Company’s potentially dilutive shares, which include outstanding share options to purchase ordinary shares, RSUs, and Series A preferred shares, are considered to be ordinary share equivalents and are only included in the calculation of diluted net loss per share when their effect is dilutive.

The following ordinary share equivalents, presented based on amounts outstanding at each period end, were excluded from the calculation of diluted net loss per share attributable to ordinary shareholders for the periods indicated because including them would have had an anti-dilutive effect:

 

 

 

As of June 30,

 

 

 

2022

 

 

2021

 

Options to purchase ordinary shares

 

 

8,526,312

 

 

 

4,584,490

 

RSUs

 

 

979,850

 

 

 

2,027,019

 

Series A preferred shares

 

 

3,901,348

 

 

 

3,901,348

 

 

Additionally, for the periods presented, the two-class method does not impact the net loss per ordinary share as the Company was in a net loss position for each of the periods presented and holders of Series A preferred shares do not participate in losses.

8. INCOME TAXES

During the three and six months ended June 30, 2022 and 2021, the Company recorded no income tax provision. The Company maintained a full valuation allowance for the three and six months ended June 30, 2022 and 2021 in all jurisdictions due to uncertainty regarding future taxable income.

The Company’s utilization of its net operating loss carryforwards and general business credit carryforwards in the United States may be subject to a substantial annual limitation under Sections 382 and 383 of the Internal Revenue Code of 1986, as amended, due to ownership changes that have occurred previously or that could occur in the future. These ownership changes may limit the amount of carryforwards that can be utilized annually to offset future taxable income. As of June 30, 2022, the Company is evaluating whether an ownership change occurred during 2022. Should an ownership change have occurred or occur in the future, the Company’s ability to utilize its net operating losses and general business credit carryforwards may be limited.

17


 

9. GEOGRAPHIC DATA

Substantially all of the Company’s long-lived assets were located in the United States as of June 30, 2022 and December 31, 2021.

10. RELATED PARTIES

The Company had the following related party transaction for the periods presented in the accompanying consolidated financial statements:

In 2012, the Company entered into a consulting agreement for scientific advisory services with Dr. Gregory L. Verdine, one of the Company’s founders and a member of the Company’s board of directors. The consulting agreement does not have a specific term and may be terminated by either party upon 14 days’ prior written notice. Pursuant to the consulting agreement, the Company pays Dr. Verdine approximately $13 thousand per month, plus reimbursement for certain expenses.

11. ACCRUED EXPENSES AND OTHER CURRENT LIABILITIES

Accrued expenses and other current liabilities consist of the following:

 

 

 

June 30, 2022

 

 

December 31, 2021

 

 

 

(in thousands)

 

Accrued compensation

 

$

6,574

 

 

$

10,181

 

Accrued expenses related to CROs and CMOs

 

 

3,473

 

 

 

3,571

 

Accrued expenses and other current liabilities

 

 

1,038

 

 

 

1,109

 

Total accrued expenses and other current liabilities

 

$

11,085

 

 

$

14,861

 

 

18


 

12. LEASES

Lease Arrangements

The Company enters into lease arrangements for its facilities. A summary of the arrangements is as follows:

Operating Leases

Lexington

On September 26, 2016, and as amended on December 31, 2016, the Company entered into a 10 year and 9-month lease , which includes two successive five-year renewal options, for its facility in Lexington, Massachusetts (the “Lexington Lease”), which the Company uses primarily for its current good manufacturing practices (“cGMP”) manufacturing, as well as for additional laboratory and office space. Throughout the term of the Lexington Lease, the Company is responsible for paying certain costs and expenses, in addition to the rent, as specified in the lease, including a proportionate share of applicable taxes, operating expenses and utilities. As required under the terms of the Lexington Lease, the Company had restricted cash of approximately $2.7 million in a separate bank account as of June 30, 2022 and December 31, 2021.

Cambridge

In April 2015, the Company entered into a lease agreement for an office and laboratory facility in Cambridge, Massachusetts (the “Cambridge Lease”), which commenced in October 2015 with a term of 7.5 years with a five-year renewal option to extend the lease. Throughout the term of the Cambridge Lease, the Company is responsible for paying certain costs and expenses, in addition to the rent, as specified in the lease, including a proportionate share of applicable taxes, operating expenses and utilities. As required under the terms of the Cambridge Lease, the Company had restricted cash of $1.0 million in a separate bank account as of June 30, 2022 and December 31, 2021.

In December 2020, the Company exercised its option under the Cambridge Lease to lease additional office and laboratory space at the adjoining facility. The combined space constitutes the entire building located at 733 Concord Avenue. The lease for the additional space commenced on October 1, 2021, with a term of five years and, for accounting purposes, is considered a separate lease from the Cambridge Lease (the “Additional Cambridge Lease”). On the commencement date of the Additional Cambridge Lease, the Company recorded a right-of-use asset and corresponding operating lease liability of $4.5 million and began recognizing straight-line rent expense under ASC 842. Throughout the term of the Additional Cambridge Lease, the Company is responsible for paying certain costs and expenses, in addition to the rent, as specified in the lease, including a proportionate share of applicable taxes, operating expenses and utilities.

In June 2022, the Company notified the landlord of its intention to exercise the five-year renewal option under the Cambridge Lease to extend the lease term through March 2028 (the “Cambridge Lease Extension”). In June 2022, the Company calculated an incremental borrowing rate of 10.53% and remeasured the right-of-use asset and the lease liabilities related to the Cambridge Lease Extension. As of June 30, 2022, the Company recorded an additional $12.0 million of operating right-of-use asset and corresponding operating lease liabilities relating to the Cambridge Lease Extension. In August 2022, the Company executed the second amendment to the Cambridge Lease with respect to the Cambridge Lease Extension.

19


 

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

The following discussion and analysis of our financial condition and results of operations should be read in conjunction with our financial statements and related notes appearing elsewhere in this Quarterly Report on Form 10-Q and in our Annual Report on Form 10-K for the year ended December 31, 2021, filed with the Securities and Exchange Commission (“SEC”) on March 3, 2022, as amended (the “2021 Annual Report on Form 10-K”). Some of the information contained in this discussion and analysis or set forth elsewhere in this Quarterly Report on Form 10-Q, including information with respect to our plans and strategy for our business, includes forward-looking statements that involve risks and uncertainties. As a result of many factors, including those factors set forth in the “Risk Factors” section of this Quarterly Report on Form 10-Q or the “Risk Factor” section of our 2021 Annual Report on Form 10-K, our actual results could differ materially from the results described in, or implied by, these forward-looking statements.

Overview

We are a clinical-stage genetic medicines company committed to delivering life-changing treatments for people battling devastating diseases. Using PRISM™, our proprietary discovery and drug development platform that enables the precise design, optimization and production of novel stereopure oligonucleotides, we aspire to develop best-in-class medicines that target the transcriptome to treat genetically defined diseases with a high degree of unmet need.

We are developing oligonucleotides that target ribonucleic acid (“RNA”) and harness existing cellular machinery to either reduce the expression of disease-promoting RNA or proteins, restore the production of functional proteins, or modulate protein expression. By intervening at the RNA level, we have the potential to address diseases that have historically been difficult to treat with small molecules or biologics, while retaining the ability to titrate dose, modulate duration of effect, and avoid risk of permanent off-target genetic changes and other challenges associated with DNA editing or gene therapy approaches. Oligonucleotides have additional advantages as a therapeutic class, including the ability to access multiple tissue types and the ability to modulate the frequency of dosing to ensure broad distribution within tissues over time. Oligonucleotides also have well-established manufacturing processes and validated test methods based on decades of improvements, as well as established regulatory, access and reimbursement pathways.

Our approach is based on the scientific insight that the biological machinery necessary to address genetic diseases already exists in human cells and can be controlled with the right tools. We have built a genetic toolkit comprised of multiple therapeutic modalities, including RNase-H mediated silencing, RNAi, splicing, and RNA base editing, all of which leverage learnings and optimizations from our PRISM platform and allow us to design built-for-purpose molecules to optimally address disease biology.

Our A-to-I(G) RNA base editing oligonucleotides (“AIMers”) represent our newest therapeutic modality. AIMers are designed to edit single base mutations on RNA transcripts, thereby avoiding permanent changes to the genome that occur with DNA-targeting approaches. Rather than using an exogenous editing enzyme, AIMers recruit proteins that exist in the body, called “ADAR” (adenosine deaminases acting on RNA) enzymes, which naturally possess the ability to change an adenine (A) to an inosine (I), which cells read as guanine (G). This approach enables simplified delivery and avoids the risk of irreversible off-target effects with DNA-targeting approaches. AIMers are short in length, fully chemically modified, and use novel chemistry, including proprietary PN backbone modifications and control of stereochemistry, which make them distinct from other ADAR-mediated editing approaches.

 

https://cdn.kscope.io/3cfc89963ab03391188fabaa3bcfe113-img55497064_0.jpg 

 

20


 

Our PRISM platform is built on the recognition that a significant opportunity exists to tune the pharmacological properties of oligonucleotide therapeutics by leveraging three key features of these molecules: sequence, chemistry, and stereochemistry. Our unique ability to control stereochemistry, which is a reality of chemically modified oligonucleotides, provides the resolution necessary to optimize pharmacological profiles.

PRISM enables us to design stereopure oligonucleotides, which are comprised of molecules with atoms precisely and purposefully arranged in three-dimensional orientations at each linkage. These differ from the mixture-based oligonucleotides currently on the market or in development by others. Additionally, to mitigate pharmacological risks and potential manufacturing challenges, our approach focuses on designing short, chemically modified oligonucleotides without the need for complex delivery vehicles or engineered exogenous enzymes.

Our work in developing stereopure oligonucleotides has enabled the continued evolution of PRISM and our drug discovery process of selecting genetically defined targets, identifying a sequence and applying the therapeutic modality we determine is best suited for the disease biology. We use our PRISM platform engine to screen candidates and optimize the pharmacologic profile based on predefined design principles, which reflect a deep understanding of how the interplay among oligonucleotide sequence, chemistry and backbone stereochemistry impacts key pharmacological properties. Through continued exploration of these interactions using iterative analysis of in vitro and in vivo outcomes and machine learning-driven predictive modeling, we also continue to refine our design principles that we deploy across subsequent programs. We continue to invest in PRISM to further evolve and apply the expanding capabilities and promise of our unique platform.

In August 2020, we publicly introduced our novel PN backbone chemistry modifications, which have been shown preclinically to increase potency, distribution, and durability of effect across various modalities. PN chemistry has been incorporated into all our current clinical, preclinical, and discovery-stage programs.

We have a robust and diverse pipeline of PN-modified, stereopure oligonucleotides, including our clinical silencing and splicing programs, as well as our AIMers. With RNA base editing, our initial focus is on using N-acetyl galactosamine (“GalNAc”)-conjugated AIMers to treat hepatic diseases. Our lead programs are designed to treat genetic diseases within the central nervous system (“CNS”), including amyotrophic lateral sclerosis (“ALS”), frontotemporal dementia (“FTD”), Huntington’s disease (“HD”), Duchenne muscular dystrophy (“DMD”), and alpha-1 antitrypsin deficiency (“AATD”). These programs include:

WVE-004 (silencing), our C9orf72-targeted variant selective antisense oligonucleotide for the treatment of C9orf72-associated ALS and FTD,
WVE-003 (silencing), our SNP3-targeted antisense oligonucleotide for the treatment of HD, which is designed to selectively lower mutant HTT protein levels, while leaving wild-type HTT protein levels relatively intact,
WVE-N531 (splicing), our exon 53 exon-skipping oligonucleotide for the treatment of DMD, and
WVE-006 (RNA editing), our GalNAc-conjugated SERPINA1 AIMer for the treatment of AATD.

When we built Wave, we recognized the growing momentum in RNA therapeutics and anticipated the value in having an internal current good manufacturing practices (“cGMP”) manufacturing facility. This capability provides us with increased control and visibility of our drug substance supply chain, thereby accelerating transitions from discovery through clinical development, and the continued ability to innovate in oligonucleotide manufacturing. Our team includes experts in oligonucleotide manufacturing that have successfully delivered clinical supply for six global studies at Wave to date. With our existing manufacturing facility, we are evaluating using our additional capacity to support the clinical supply of innovative oligonucleotides for new partners.

21


 

Our Current Programs

 

https://cdn.kscope.io/3cfc89963ab03391188fabaa3bcfe113-img55497064_1.jpg 

 

Additional details regarding our lead therapeutic programs are set forth below.

Neurology

WVE-004: In ALS and FTD, we are advancing WVE-004, which uses our novel PN chemistry and preferentially targets the transcripts containing the hexanucleotide G4C2 expansion in the C9orf72 gene. In C9 BAC transgenic mice, WVE-004 led to substantial reductions in repeat-containing C9orf72 transcripts and dipeptide repeat (“DPR”) proteins that are sustained for at least six months, without disrupting total C9orf72 protein expression.

In 2021, we initiated our FOCUS-C9 trial, which is a global, multicenter, randomized, double-blind, placebo-controlled Phase 1b/2a clinical trial to assess the safety and tolerability of intrathecal doses of WVE-004 for patients with C9-ALS and/or C9-FTD. Additional objectives include measurement of polyGP proteins in the cerebrospinal fluid (“CSF”), plasma and CSF pharmacokinetics and exploratory biomarker and clinical endpoints. The FOCUS-C9 trial is designed to be adaptive with dose level and dosing frequency being guided by an independent committee. Preclinical models that have established pharmacologic activity have informed the starting dose for this trial.

22


 

In April 2022, we announced a positive update to the ongoing FOCUS-C9 trial that was driven by the observation of potent, durable reductions of poly(GP) dipeptide repeat proteins in CSF with low, single doses of WVE-004. Poly(GP) is a key C9-ALS/C9-FTD disease biomarker that, when reduced in CSF, indicates WVE-004’s engagement of target in the brain and spinal cord. Poly(GP) is also a DPR protein translated from both sense and antisense transcripts of the C9orf72 repeat expansions responsible for the pathophysiology of disease.

Reductions in poly(GP) were observed across all active treatment groups (10 mg, n=2 patients; 30 mg, n=4 patients; 60 mg, n=3 patients), reaching statistical significance versus placebo (n=3 patients) after single 30 mg doses, with a 34% reduction in poly(GP) at day 85 (p=0.011). At the time of analysis, none of the patients dosed with 60 mg had reached day 85.

 

https://cdn.kscope.io/3cfc89963ab03391188fabaa3bcfe113-img55497064_2.jpg 

 

As the poly(GP) reduction in the 30 mg single dose cohort does not appear to have plateaued, we are extending the observation period from approximately three months (85 days) to approximately six months to identify the maximum reduction of poly(GP) and duration of effect of low single doses. Based on the durability and potency observed in the 30 mg cohort, FOCUS-C9 has been adapted to include additional patients receiving 20 mg and 30 mg single doses of WVE-004.
Additional exploratory assessments included monitoring of CSF neurofilament light chain (“NfL”) and clinical outcome measures. CSF NfL elevations were observed in some patients in the 30 mg and 60 mg single dose cohorts with no meaningful changes in clinical outcome measures, although the dataset and duration were not sufficient to assess clinical effects. Exploratory assessments will continue throughout the single and multidose phases of the FOCUS-C9 trial.
Adverse events were balanced across treatment groups, including placebo, and were mostly mild to moderate in intensity. Four patients (including one on placebo) experienced severe and/or serious adverse events; three were reported by the investigators to be related to ALS or administration, and one was reported by the investigator to be related to study drug. There were no treatment-associated elevations in CSF white blood cell counts or protein and no other notable laboratory abnormalities were observed.

We expect to share additional single and multidose data from the FOCUS-C9 clinical trial in the second half of 2022. We expect to use these data to optimize dose level and frequency, as well as enable discussions with regulatory authorities regarding the next phase of development later in 2022. Planning is underway for expected initiation of an open-label extension clinical trial in the second half of 2022.

23


 

WVE-003: In HD, we are currently advancing WVE-003, a stereopure antisense oligonucleotide designed to selectively target an undisclosed single nucleotide polymorphism (“SNP”), “mHTT SNP3”, associated with the disease-causing mutant huntingtin (“mHTT”) mRNA transcript within the HTT gene. WVE-003 incorporates our novel PN chemistry, as well as learnings from our first-generation HD programs. Targeting mRNA with SNP3 allows us to lower expression of transcript from the mutant allele, while leaving the healthy transcript relatively intact, thereby preserving wild-type (healthy) huntingtin (“wtHTT”) protein, which is important for neuronal function. Our allele-selective approach may also enable us to address the pre-manifest, or asymptomatic, HD patient population in the future. In preclinical studies, WVE-003 showed dose-dependent and selective reduction of mHTT mRNA in vitro and potent and durable knockdown of mHTT mRNA and protein in vivo. A pharmacokinetic-pharmacodynamic (“PK-PD”) model for WVE-003 based on preclinical data predicts that WVE-003 may attain sufficient concentrations to engage mHTT transcript in both the cortex and striatum and decrease expression of the mHTT protein.

The SELECT-HD trial is a multicenter, randomized, double-blind, placebo-controlled Phase 1b/2a clinical trial to assess the safety and tolerability of intrathecally administered WVE-003 for patients with early manifest HD. Additional objectives include measurement of mHTT and wtHTT protein and exploratory pharmacokinetic, pharmacodynamic, clinical and magnetic resonance imaging (“MRI”) endpoints. The SELECT-HD trial is designed to be adaptive, with dose level and dosing frequency being guided by an independent committee. Preclinical models that have established pharmacologic activity have informed the starting dose for this trial. In September 2021, we announced the initiation of dosing in the SELECT-HD trial. We expect to share clinical data in the second half of 2022 to provide further insight into the clinical effects of PN chemistry and enable decision making for WVE-003.

WVE-N531: In DMD, we are advancing WVE-N531, which is designed to target exon 53 within the dystrophin gene. WVE-N531 is designed to cause the cellular splicing machinery to skip over this exon during pre-mRNA processing, which restores the dystrophin mRNA reading frame and enables production of truncated, but functional dystrophin protein. Exon-skipping produces dystrophin from the endogenous dystrophin gene (not micro or mini dystrophin expressed from a vector), under the control of native gene-regulatory elements, resulting in normal temporospatial expression. WVE-N531 is both our first splicing candidate and our first systemically administered candidate incorporating PN chemistry to be assessed in the clinic.

In September 2021, we announced the initiation of dosing in an open-label clinical trial evaluating WVE-N531 as a treatment for boys with DMD who are amenable to exon 53 skipping. We expect to share clinical data, including muscle biopsies, in the fourth quarter of 2022 to provide further insight into the clinical effects of PN chemistry and enable decision making for WVE-N531.

Hepatic

We are initially focused on developing AIMers (A-to-I(G) RNA base editing oligonucleotides), which use our novel PN chemistry, to address genetic hepatic diseases. Our AIMers are relatively short and stable (fully chemically modified), which enables us to leverage clinically-proven GalNAc-mediated delivery to hepatocytes with subcutaneous dosing.

WVE-006: In AATD, we are currently advancing WVE-006, our first AIMer (RNA editing) candidate that uses GalNAc-conjugation and endogenous ADAR enzymes to correct the SERPINA1 Z mutation, which is the most common cause of AATD. ADAR editing may provide an ideal approach for increasing circulating levels of wild-type AAT protein and reducing aggregation in the liver, thus simultaneously addressing both the lung and liver manifestations of the disease. We selected WVE-006 as our AATD AIMer development candidate, our IND-enabling activities are underway, and we expect to submit clinical trial applications in 2023.

Continuing Impacts of COVID-19

We continue to closely monitor developments related to COVID-19, which was declared a pandemic by the World Health Organization on March 11, 2020. In response to this global pandemic, we have concentrated our efforts on the health and safety of our employees and patients, while maintaining business continuity and honoring our commitment to deliver life-changing treatments for people battling devastating diseases.

Our on-site activities continue with protocols for safely accessing and working within our facilities, including a mandatory COVID-19 vaccination policy that we implemented in October 2021. While we continue to conduct research and development activities, including our ongoing clinical trials, the COVID-19 pandemic has impacted, and may continue to impact, certain of our early-stage discovery efforts and clinical trials. We are working with our clinical investigators, research and development vendors, and supply chain vendors to continually assess and take steps to mitigate the potential impact of COVID-19 on our manufacturing operations and research and development activities.

We will continue to closely monitor the COVID-19 situation as we evolve our business continuity plans. Given the global risks and uncertainties associated with COVID-19, our business, results of operations, and prospects could be materially adversely affected. For additional information, see “Item 1A. Risk Factors” in the 2021 Annual Report on Form 10-K.

24


 

Financial Operations Overview

We have never been profitable, and since our inception, we have incurred significant operating losses. Our net loss was $41.3 million and $38.8 million for the three months ended June 30, 2022 and 2021, respectively. Our net loss was $79.1 million and $81.2 million for the six months ended June 30, 2022 and 2021, respectively. As of June 30, 2022 and December 31, 2021, we had an accumulated deficit of $884.6 million and $805.5 million, respectively. We expect to incur significant expenses and operating losses for the foreseeable future.

Revenue

We have not generated any product revenue since our inception and do not expect to generate any revenue from the sale of products for the foreseeable future. Under the revenue recognition standard, we recognize collaboration revenue under the Takeda Collaboration Agreement (as defined in Note 5 in the notes to the unaudited consolidated financial statements appearing elsewhere in this Quarterly Report on Form 10-Q, “Note 5”), which became effective in April 2018.

Operating Expenses

Our operating expenses since inception have consisted primarily of research and development expenses and general and administrative expenses.

Research and Development Expenses

Research and development expenses consist primarily of costs incurred for our research activities, including our discovery efforts, and the development of our product candidates, which include:

compensation-related expenses, including employee salaries, bonuses, share-based compensation expense and other related benefits expenses for personnel in our research and development organization;
expenses incurred under agreements with third parties, including contract research organizations (“CROs”) that conduct research, preclinical and clinical activities on our behalf, as well as contract manufacturing organizations (“CMOs”) that manufacture drug product for use in our preclinical studies and clinical trials;
expenses incurred related to our internal manufacturing of drug substance for use in our preclinical studies and clinical trials;
expenses related to compliance with regulatory requirements;
expenses related to third-party consultants;
research and development supplies and services expenses; and
facility-related expenses, including rent, maintenance and other general operating expenses.

We recognize research and development costs as incurred. We recognize external development costs based on an evaluation of the progress to completion of specific tasks using information provided to us by our vendors. Payments for these activities are based on the terms of the individual agreements, which may differ from the pattern of costs incurred, and are reflected in our financial statements as prepaid or accrued expenses.

Our primary research and development focus since inception has been the development of our proprietary discovery and drug development platform, PRISM. We are using PRISM, which includes our novel PN backbone chemistry modifications, to design, develop and commercialize a broad pipeline of nucleic acid therapeutic candidates that target RNA using silencing, splicing, and ADAR editing.

25


 

Our research and development expenses consist primarily of expenses related to our CROs, CMOs, consultants, other external vendors and fees paid to global regulatory agencies to conduct our clinical trials, in addition to compensation-related expenses, internal manufacturing expenses, facility-related expenses and other general operating expenses. These expenses are incurred in connection with research and development efforts and our preclinical studies and clinical trials. We track certain external expenses on a program-by-program basis. However, we do not allocate compensation-related expenses, internal manufacturing expenses, equipment repairs and maintenance expense, facility-related expenses or other operating expenses to specific programs. These expenses, which are not allocated on a program-by-program basis, are included in the “PRISM and other research and development expenses” category along with other external expenses related to our discovery and development programs, as well as platform development and identification of potential drug discovery candidates.

Product candidates in later stages of clinical development generally have higher development costs than those in earlier stages of clinical development, primarily due to the increased size and duration of later-stage clinical trials. We expect to continue to incur significant research and development expenses in the foreseeable future as we continue to manage our existing clinical trials, initiate additional clinical trials for certain product candidates, pursue later stages of clinical development for certain product candidates, maintain our manufacturing capabilities and continue to discover and develop additional product candidates in multiple therapeutic areas.

General and Administrative Expenses

General and administrative expenses consist primarily of compensation-related expenses, including salaries, bonuses, share-based compensation and other related benefits costs for personnel in our executive, finance, corporate, legal and administrative functions, as well as compensation-related expenses for our board of directors. General and administrative expenses also include legal fees; expenses associated with being a public company; professional fees for accounting, auditing, tax and consulting services; insurance costs; travel expenses; other operating costs; and facility-related expenses.

Other Income, Net

Other income, net consists primarily of refundable tax credits from tax authorities. We recognize refundable tax credits when there is reasonable assurance that we will comply with the requirements of the refundable tax credit and that the refundable tax credit will be received.

Income Taxes

We are a Singapore multi-national company subject to taxation in the United States and various other jurisdictions.

Critical Accounting Policies and Significant Judgments and Estimates

Our consolidated financial statements are prepared in accordance with generally accepted accounting principles in the United States. The preparation of our financial statements and related disclosures requires us to make estimates and assumptions that affect the reported amount of assets, liabilities, revenue, costs and expenses and related disclosures.

Our significant accounting policies, judgments and estimates are described in Note 2 in the notes to the audited consolidated financial statements included in the 2021 Annual Report on Form 10-K, as well as in Note 2 in the notes to the unaudited consolidated financial statements included in this Quarterly Report on Form 10-Q. We believe that our revenue recognition policy, particularly (a) assessing the number of performance obligations; (b) determining the transaction price; (c) allocating the transaction price to the performance obligations in the contract; and (d) determining the pattern over which performance obligations are satisfied, including estimates to complete performance obligations, and the assumptions and estimates used in our analysis of contracts with CROs and CMOs to estimate the contract expense, involve a greater degree of judgment, and therefore we consider them to be our critical accounting policies. We evaluate our estimates and assumptions on an ongoing basis. Our actual results may differ from these estimates under different assumptions and conditions.

26


 

Results of Operations

Comparison of the three months ended June 30, 2022 and 2021

 

 

 

 

Three Months Ended June 30,

 

 

 

 

 

 

2022

 

 

2021

 

 

Change

 

 

 

(in thousands)

 

Revenue

 

$

375

 

 

$

2,776

 

 

$

(2,401

)

Operating expenses:

 

 

 

 

 

 

 

 

 

Research and development

 

 

29,733

 

 

 

31,635

 

 

 

(1,902

)

General and administrative

 

 

12,806

 

 

 

10,969

 

 

 

1,837

 

Total operating expenses

 

 

42,539

 

 

 

42,604

 

 

 

(65

)

Loss from operations

 

 

(42,164

)

 

 

(39,828

)

 

 

(2,336

)

Total other income, net

 

 

868

 

 

 

1,062

 

 

 

(194

)

Loss before income taxes

 

 

(41,296

)

 

 

(38,766

)

 

 

(2,530

)

Income tax provision

 

 

 

 

 

 

 

 

 

Net loss

 

$

(41,296

)

 

$

(38,766

)

 

$

(2,530

)

 

Revenue

Revenue for the three months ended June 30, 2022 and 2021 was $0.4 million and $2.8 million, respectively, and was primarily earned under the Takeda Collaboration Agreement. The decrease in revenue is driven by an increase in the expected future research and development services under the Takeda Collaboration Agreement based on the revenue recognition standard, as well as the discontinuation of the Category 2 component in the third quarter of 2021.

Research and Development Expenses

 

 

 

Three Months Ended June 30,

 

 

 

 

 

 

2022

 

 

2021

 

 

Change

 

 

 

(in thousands)

 

ALS and FTD program

 

$

3,348

 

 

$

3,381

 

 

$

(33

)

HD programs

 

 

1,253

 

 

 

6,911

 

 

 

(5,658

)

DMD programs

 

 

392

 

 

 

356

 

 

 

36

 

AATD program

 

 

1,541

 

 

 

3

 

 

 

1,538

 

PRISM and other research and development expenses (1)

 

 

23,199

 

 

 

20,984

 

 

 

2,215

 

Total research and development expenses

 

$

29,733

 

 

$

31,635

 

 

$

(1,902

)

 

(1)
Includes expenses related to discovery and development programs, identification of potential drug discovery candidates, compensation, internal manufacturing, equipment repairs and maintenance, facilities and other operating expenses, which are not allocated to specific programs.

Research and development expenses were $29.7 million for the three months ended June 30, 2022, compared to approximately $31.6 million for the three months ended June 30, 2021. The decrease of $1.9 million was due to the following:

a decrease of less than $0.1 million in external expenses related to our ALS and FTD program, WVE-004 (PN-modified silencing oligonucleotide);
a decrease of $5.7 million in external expenses related to our HD programs, driven by decreased external expenses related to our discontinued WVE-120101 and WVE-120102 programs, partially offset by continuing external expenses for our WVE-003 (PN-modified silencing oligonucleotide) program;
an increase of less than $0.1 million in external expenses related to our DMD programs, including WVE-N531 (PN-modified splicing oligonucleotide);
an increase of $1.5 million in external expenses related to our AATD program, WVE-006 (PN-modified RNA editing oligonucleotide); and
an increase of $2.2 million in internal and external research and development expenses that are not allocated on a program-by-program basis and are related to other discovery and development programs, including PRISM and the identification of potential drug discovery candidates, primarily due to increases in compensation-related expenses,

27


 

including the share-based compensation expense related to the achievement of a performance-based RSU milestone, offset by decreases in other external research and development expenses.

General and Administrative Expenses

General and administrative expenses were $12.8 million for the three months ended June 30, 2022, as compared to $11.0 million for the three months ended June 30, 2021. The increase of $1.8 million is primarily driven by increases in compensation-related expenses, including the share-based compensation expense related to the achievement of a performance-based RSU milestone, offset by decreases in other external general and administrative expenses.

Other Income, Net

Other income, net for the three months ended June 30, 2022 and 2021, was $0.9 million and $1.1 million, respectively. Other income, net for both periods primarily consisted of income related to refundable tax credits from tax authorities.

Income Tax Provision

During the three months ended June 30, 2022 and 2021, we recorded no income tax provision. We maintained a full valuation allowance for the three months ended June 30, 2022 and 2021 in all jurisdictions due to uncertainty regarding future taxable income.

Comparison of the six months ended June 30, 2022 and 2021

 

 

 

Six Months Ended June 30,

 

 

 

 

 

 

2022

 

 

2021

 

 

Change

 

 

 

(in thousands)

 

Revenue

 

$

2,125

 

 

$

2,776

 

 

$

(651

)

Operating expenses:

 

 

 

 

 

 

 

 

 

Research and development

 

 

57,203

 

 

 

65,028

 

 

 

(7,825

)

General and administrative

 

 

25,180

 

 

 

21,047

 

 

 

4,133

 

Total operating expenses

 

 

82,383

 

 

 

86,075

 

 

 

(3,692

)

Loss from operations

 

 

(80,258

)

 

 

(83,299

)

 

 

3,041

 

Total other income, net

 

 

1,148

 

 

 

2,069

 

 

 

(921

)

Loss before income taxes

 

 

(79,110

)

 

 

(81,230

)

 

 

2,120

 

Income tax provision

 

 

 

 

 

 

 

 

 

Net loss

 

$

(79,110

)

 

$

(81,230

)

 

$

2,120

 

 

Revenue

Revenue for the six months ended June 30, 2022 and 2021 was $2.1 million and $2.8 million, respectively, and was primarily earned under the Takeda Collaboration Agreement. The decrease in revenue is driven by the discontinuation of the Category 2 component in the third quarter of 2021.

28


 

Research and Development Expenses

 

 

 

Six Months Ended June 30,

 

 

 

 

 

 

2022

 

 

2021

 

 

Change

 

 

 

(in thousands)

 

ALS and FTD program

 

$

5,087

 

 

$

6,821

 

 

$

(1,734

)

HD programs

 

 

3,446

 

 

 

15,361

 

 

 

(11,915

)

DMD programs

 

 

817

 

 

 

156

 

 

 

661

 

AATD program

 

 

1,852

 

 

 

3

 

 

 

1,849

 

PRISM and other research and development expenses (1)

 

 

46,001

 

 

 

42,687

 

 

 

3,314

 

Total research and development expenses

 

$

57,203

 

 

$

65,028

 

 

$

(7,825

)

 

(1)
Includes expenses related to discovery and development programs, identification of potential drug discovery candidates, compensation, internal manufacturing, equipment repairs and maintenance, facilities and other operating expenses, which are not allocated to specific programs.

Research and development expenses were $57.2 million for the six months ended June 30, 2022, compared to $65.0 million for the six months ended June 30, 2021. The decrease of $7.8 million was due to the following:

a decrease of $1.7 million in external expenses related to our ALS and FTD program, WVE-004 (PN-modified silencing oligonucleotide);
a decrease of $11.9 million in external expenses related to our HD programs, driven by decreased external expenses related to our discontinued WVE-120101 and WVE-120102 programs, partially offset by continuing external expenses for our WVE-003 (PN-modified silencing oligonucleotide) program;
an increase of $0.7 million in external expenses related to our DMD programs, including WVE-N531 (PN-modified splicing oligonucleotide);
an increase of $1.8 million in external expenses related to our AATD program, WVE-006 (PN-modified RNA editing oligonucleotide); and
an increase of $3.3 million in internal and external research and development expenses that are not allocated on a program-by-program basis and are related to other discovery and development programs, including PRISM and the identification of potential drug discovery candidates, primarily due to increases in compensation-related expenses, including the share-based compensation expense related to the achievement of a performance-based RSU milestone.

General and Administrative Expenses

General and administrative expenses were $25.2 million for the six months ended June 30, 2022, as compared to approximately $21.0 million for the six months ended June 30, 2021. The increase of $4.1 million was primarily due to increases in compensation-related expenses, including the share-based compensation expense related to the achievement of a performance-based RSU milestone, as well as increases in professional services expenses and other general and administrative operating expenses.

Other Income, Net

Other income, net for the six months ended June 30, 2022 and 2021, was $1.1 million and $2.1 million, respectively. Other income, net for both periods primarily consisted of income related to refundable tax credits from tax authorities.

Income Tax Provision

During the six months ended June 30, 2022 and 2021, we recorded no income tax provision. We maintained a full valuation allowance for the six months ended June 30, 2022 and 2021 in all jurisdictions due to uncertainty regarding future taxable income.

29


 

Liquidity and Capital Resources

Since our inception, we have not generated any product revenue and have incurred recurring net losses. To date, we have primarily funded our operations through public and other registered offerings of our ordinary shares, collaborations with third parties and private placements of debt and equity securities. Through June 30, 2022, we have received an aggregate of approximately $1,021.2 million in net proceeds from these transactions, consisting of $630.9 million in net proceeds from public and other registered offerings of our ordinary shares, $301.0 million from our collaborations and $89.3 million in net proceeds from private placements of our debt and equity securities.

On June 16, 2022, we closed an underwritten offering (the “June 2022 Offering”), in which we issued and sold 25,464,483 ordinary shares and Pre-Funded Warrants to purchase up to 7,093,656 ordinary shares. The net proceeds of the June 2022 Offering were approximately $65.5 million.

As of June 30, 2022, we had cash, cash equivalents and short-term investments totaling $148.2 million, restricted cash of $3.7 million for our leased premises in Cambridge, Massachusetts and Lexington, Massachusetts, and an accumulated deficit of $884.6 million. Our operating lease commitments as of June 30, 2022 total approximately $50.6 million, of which $3.6 million is related to payments in 2022 and $47.0 million is related to payments beyond 2022.

We expect that our existing cash, cash equivalents and short-term investments will be sufficient to fund our operations for at least the next twelve months. We have based this expectation on assumptions that may prove to be incorrect, and we may use our available capital resources sooner than we currently expect. In addition, we may elect to raise additional funds before we need them if the conditions for raising capital are favorable due to market conditions or strategic considerations, even if we expect we have sufficient funds for our current or future operating plans.

Until we can generate significant revenue from product sales, if ever, we expect to continue to finance our operations through a combination of public or private equity or debt financings or other sources, which may include collaborations with third parties. In May 2019, we filed a shelf registration statement on Form S-3ASR with the SEC pursuant to which we registered for sale an indeterminate amount of any combination of our ordinary shares, debt securities, warrants, rights and/or units from time to time and at prices and on terms that we may determine. Our shelf registration statement on Form S-3ASR also included a prospectus covering up to an aggregate of $250.0 million in ordinary shares that we may issue and sell from time to time, through Jefferies LLC (“Jefferies”) acting as our sales agent, pursuant to the open market sales agreement that we entered into with Jefferies in May 2019, as amended in March 2020 and March 2022 (the “Sales Agreement”), for our “at-the-market” equity program. Since we no longer qualified as a “well-known seasoned issuer” at the time of the filing of our Annual Report on Form 10-K for the year ended December 31, 2019, we previously amended the shelf registration statement to register for sale up to $500.0 million of any combination of our ordinary shares, debt securities, warrants, rights and/or units from time to time and at prices and on terms that we may determine, including the $250.0 million in ordinary shares that we may issue and sell from time to time pursuant to our “at-the-market” equity program. This registration statement, which we refer to as the “2019 Form S-3,” remained effective until our 2022 Form S-3 (as defined below) was declared effective on May 4, 2022, after which time we may no longer offer or sell any securities under the 2019 Form S-3. During the six months ended June 30, 2022, the Company sold 458,092 ordinary shares under its at-the-market equity program for aggregate net proceeds of $1.2 million.

On March 3, 2022, we filed a new universal shelf registration on Form S-3 with the SEC, which was declared effective by the SEC on May 4, 2022, pursuant to which we registered for sale up to $500.0 million of any combination of our ordinary shares, debt securities, warrants, rights and/or units from time to time and at prices and on terms that we may determine, which we refer to as the “2022 Form S-3.” The 2022 Form S-3 includes a prospectus covering up to approximately $132.0 million in ordinary shares that have not yet been issued or sold under our Sales Agreement with Jefferies. As of August 10, 2022, we have $430.0 million in securities available for issuance under the 2022 Form S-3, including approximately $132.0 million in ordinary shares available for issuance under our at-the-market equity program.

Adequate additional financing may not be available to us on acceptable terms, or at all. Our inability to raise capital as and when needed would have a negative impact on our financial condition and our ability to pursue our business strategy. We will need to generate significant revenue to achieve profitability, and we may never do so.

30


 

Cash Flows

The following table summarizes our cash flow activity:

 

 

 

Six Months Ended June 30,

 

 

 

2022

 

 

2021

 

 

 

(in thousands)

 

Net cash used in operating activities

 

$

(68,695

)

 

$

(53,776

)

Net cash used in investing activities

 

 

(25,594

)

 

 

(447

)

Net cash provided by financing activities

 

 

67,116

 

 

 

13,686

 

Effect of foreign exchange rates on cash, cash equivalents and restricted cash

 

 

(228

)

 

 

(120

)

Net decrease in cash, cash equivalents and restricted cash

 

$

(27,401

)

 

$

(40,657

)

 

Operating Activities

During the six months ended June 30, 2022, operating activities used $68.7 million of cash, primarily due to our net loss of $79.1 million, offset by $10.9 million of share-based compensation expense.

During the six months ended June 30, 2021, operating activities used $53.8 million of cash, primarily due to our net loss of $81.2 million, offset by a $30.0 million decrease in accounts receivable.

Investing Activities

During the six months ended June 30, 2022, investing activities used $25.6 million of cash, of which $50.0 million related to purchases of short-term investments, $25.0 million of which was subsequently sold and $0.6 million related to purchases and sales of property and equipment.

During the six months ended June 30, 2021, investing activities used $0.4 million of cash, related to purchases of property and equipment.

Financing Activities

During the six months ended June 30, 2022, net cash provided by financing activities was $67.1 million, which was primarily due to the net proceeds from the June 2022 Offering, which was comprised of sales of ordinary shares and Pre-Funded Warrants.

During the six months ended June 30, 2021, net cash provided by financing activities was $13.7 million, which was primarily due to the net proceeds from sales of ordinary shares under our at-the-market equity program.

Funding Requirements

We expect to continue to incur significant expenses in connection with our ongoing research and development activities and our internal cGMP manufacturing activities. Furthermore, we anticipate that our expenses will continue to vary if and as we:

continue to conduct our clinical trials evaluating our product candidates in patients;
conduct research and preclinical development of discovery targets and advance additional programs into clinical development;
file clinical trial applications with global regulatory agencies and conduct clinical trials for our programs;
evaluate next steps for our programs in rare, inherited eye diseases;
make strategic investments in continuing to innovate our research and development platform, PRISM, and in optimizing our manufacturing processes and formulations;
maintain our manufacturing capabilities through our internal facility and our CMOs;
maintain our intellectual property portfolio and consider the acquisition of complementary intellectual property;
seek and obtain regulatory approvals for our product candidates;
respond to the impacts of the COVID-19 global pandemic on our business; and
establish and build capabilities to market, distribute and sell our product candidates.

31


 

We may experience delays or encounter issues with any of the above, including but not limited to failed studies, complex results, safety issues or other regulatory challenges.

Because of the numerous risks and uncertainties associated with the development of drug candidates and because the extent to which we may enter into collaborations with third parties for development of product candidates is unknown, we are unable to estimate the amounts of future capital outlays and operating expenses associated with completing the research and development for our therapeutic programs. Our future capital requirements for our therapeutic programs will depend on many factors, including:

the progress, results and costs of conducting research and continued preclinical and clinical development for our therapeutic programs and future potential pipeline candidates;
the number and characteristics of product candidates and programs that we pursue;
the cost of manufacturing clinical supplies of our product candidates;
whether and to what extent milestone events are achieved under our collaboration with Takeda or any potential future licensee or collaborator;
the costs, timing and outcome of regulatory review of our product candidates;
our ability to obtain marketing approval for our product candidates;
the impacts of the COVID-19 global pandemic on our business;
the costs and timing of future commercialization activities, including manufacturing, marketing, sales and distribution, for any of our product candidates for which we receive marketing approval;
market acceptance of our product candidates, to the extent any are approved for commercial sale, and the revenue, if any, received from commercial sales of our product candidates for which we receive marketing approval;
the costs and timing of preparing, filing and prosecuting patent applications, maintaining and enforcing our intellectual property rights and defending any intellectual property-related claims;
the effect of competing technological and market developments; and
the extent to which we acquire or invest in businesses, products and technologies, including entering into licensing or collaboration arrangements for product candidates.

Identifying potential product candidates and conducting preclinical testing and clinical trials is a time-consuming, expensive and uncertain process that takes years to complete, and we may never generate the necessary data or results required to obtain marketing approval and achieve product sales. In addition, our product candidates, if approved, may not achieve commercial success. Our product revenue, if any, will be derived from sales of products that we do not expect to be commercially available for many years, if ever. Accordingly, we will need to obtain substantial additional funds to achieve our business objectives.

Adequate additional funds may not be available to us on acceptable terms when we need them, or at all. We do not currently have any committed external source of funds, except for possible future payments from Takeda under the Takeda Collaboration Agreement. To the extent that we raise additional capital through the sale of equity or convertible debt securities, the ownership interest of our existing shareholders will be diluted, and the terms may include liquidation or other preferences that adversely affect the rights of our shareholders. Additional debt financing and preferred equity financing, if available, may involve agreements that include covenants limiting or restricting our ability to take specific actions, such as incurring additional debt, making capital expenditures or declaring dividends and may require the issuance of warrants, which could potentially dilute our shareholders’ ownership interests.

If we raise additional funds through collaborations, strategic alliances or licensing arrangements with third parties, we may have to relinquish valuable rights to our technologies, future revenue streams, research programs 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 programs or any future commercialization efforts or grant rights to develop and market product candidates that we would otherwise prefer to develop and market ourselves.

32


 

Item 3. Quantitative and Qualitative Disclosures About Market Risk

Market risk represents the risk of loss that may impact our financial position due to adverse changes in financial market prices and rates. Our market risk exposure is primarily the result of fluctuations in interest rates and foreign exchange rates, as well as, to a lesser extent, inflation and capital market risk.

Interest Rate Risk

We are exposed to interest rate risk in the ordinary course of our business. Our cash and cash equivalents are held in readily available checking and money market accounts. Our short-term investments are comprised of term deposits which have fixed interest rates.

Foreign Currency Exchange Rate Risk

Due to our operations outside of the United States, we are exposed to market risk related to changes in foreign currency exchange rates. Historically, we have not hedged our foreign currency exposure. For the three and six months ended June 30, 2022 and 2021, changes in foreign currency exchange rates did not have a material impact on our business, financial condition, results of operations or cash flows.

Inflation Risk

We do not believe that inflation had a material effect on our business, financial condition, results of operations or cash flows for the three and six months ended June 30, 2022 and 2021. If the global inflationary trends continue, we expect appreciable increases in clinical trial, labor, and other operating costs.

Capital Market Risk

We currently have no product revenues and depend on funds raised through other sources. One possible source of funding is through further equity offerings. Our ability to raise funds in this manner depends upon capital market forces affecting our share price, including impacts of the COVID-19 pandemic on the capital markets.

Item 4. Controls and Procedures

Evaluation of Disclosure Controls and Procedures

Our management, with the participation of our principal executive officer and principal financial officer, evaluated the effectiveness of our disclosure controls and procedures as of June 30, 2022. The term “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”), means controls and other procedures of a company that are designed to ensure that information required to be disclosed by a company in the reports that it files or submits under the Exchange Act is recorded, processed, summarized and reported, within the time periods specified in the SEC’s rules and forms. Disclosure controls and procedures include, without limitation, controls and procedures designed to ensure that information required to be disclosed by a company in the reports that it files or submits under the Exchange Act is accumulated and communicated to its management, including its principal executive and principal financial officers, as appropriate to allow timely decisions regarding required disclosure. Management recognizes that any controls and procedures, no matter how well designed and operated, can provide only reasonable assurance of achieving their objectives and management necessarily applies its judgment in evaluating the cost-benefit relationship of possible controls and procedures. Based on the evaluation of our disclosure controls and procedures as of June 30, 2022, our principal executive officer and principal financial officer concluded that, as of such date, our disclosure controls and procedures were effective at the reasonable assurance level.

Changes in Internal Control over Financial Reporting

There were no changes in our internal control over financial reporting identified in connection with the evaluation of such internal control required by Rule 13a-15(d) and 15d-15(d) of the Exchange Act that occurred during the three months ended June 30, 2022 that have materially affected, or are reasonably likely to materially affect, our internal control over financial reporting.

33


 

PART II – OTHER INFORMATION

We are not currently a party to any material legal proceedings.

Item 1A. Risk Factors

In addition to the other information set forth in this Quarterly Report on Form 10-Q, including the risk factor below, you should carefully consider the factors discussed under the caption “Risk Factors” that appear in Item 1A of our 2021 Annual Report on Form 10-K.

We issued pre-funded warrants as part of our June 2022 financing, which may cause additional dilution to our shareholders.

In June 2022, we closed an underwritten offering in which we issued and sold 25,464,483 ordinary shares and, to RA Capital Management, L.P. in lieu of additional ordinary shares, pre-funded warrants (“Pre-Funded Warrants”) to purchase up to 7,093,656 ordinary shares at an exercise price of $0.0001 per share. The Pre-Funded Warrants contain a so-called “blocker” provision which provides that they are only exercisable upon receipt of shareholder approval or if such exercise would not cause the aggregate number of ordinary shares or the combined voting power of total securities, in each case, beneficially owned by the holder (together with its affiliates) to exceed 19.99% of the number of ordinary shares or total securities, respectively, outstanding immediately after giving effect to the exercise. To the extent the Pre-Funded Warrants above are exercised, additional ordinary shares will be issued and such issuance would dilute existing shareholders and increase the number of shares eligible for resale in the public market.
 

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

Recent Sales of Unregistered Equity Securities

None.

Issuer Purchases of Equity Securities

We did not repurchase any of our equity securities during the three months ended June 30, 2022.

Item 3. Defaults Upon Senior Securities

None.

Item 4. Mine Safety Disclosures

Not applicable.

Item 5. Other Information

The information set forth in this paragraph is included herein for the purpose of providing the disclosure required under “Item 1.01 - Entry into a Material Definitive Agreement” of Form 8-K. On August 8, 2022, Wave Life Sciences USA, Inc. (the “Subsidiary”), a wholly-owned subsidiary of Wave Life Sciences Ltd. (and together with the Subsidiary, the “Company”), entered into the Second Amendment to its lease with CPI/King 733 Concord Owner, LLC (the “Second Lease Amendment”), which, among other things, extended the term of the lease at 733 Concord Avenue, Cambridge, Massachusetts, which serves as the Company’s U.S. headquarters. The Second Lease Amendment amends the lease agreement dated as of April 6, 2015, between the Subsidiary and King 733 Concord LLC, an affiliate of King Street Properties Investments, LLC (the “Original Lease”), as amended by the First Amendment to the Original Lease dated December 9, 2020. Under the Second Lease Amendment, the lease term with respect to the approximately 30,893 square feet of space leased under the Original Lease (the “Original Premises”) was extended for an additional period of five years commencing on April 1, 2023, and expiring as of March 31, 2028. During the extension period, the Company has agreed to pay an average base rent for the Original Premises of approximately $3.3 million per year, plus certain costs and expenses, including a proportionate share of applicable taxes, operating expenses and utilities. In addition, the Company received a tenant improvement allowance of up to $0.9 million. The foregoing description of the Second Lease Amendment does not purport to be complete and is qualified in its entirety by reference to the full text of the Second Lease Amendment, a copy of which is attached as an exhibit this Quarterly Report on Form 10-Q.

34


 

Item 6. Exhibits

 

Exhibit

Number

 

Exhibit Description

 

Filed

with this

Report

 

Incorporated by

Reference herein

from Form or

Schedule

 

Filing

Date

 

SEC

File/Reg.

Number

 

 

 

 

 

 

 

 

 

 

 

4.1

 

Form of Pre-Funded Warrant

 

 

 

Form 8-K

(Exhibit 4.1)

 

06/14/2022

 

001-37627

 

 

 

 

 

 

 

 

 

 

 

10.1

 

Second Amendment (to Lease) by and between Wave Life Sciences USA, Inc. and CPI/King 733 Concord Owner, LLC, dated as of August 8, 2022

 

X

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

31.1

 

Rule 13a-14(a)/15d-14(a) Certification of Principal Executive Officer

 

X

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

31.2

 

Rule 13a-14(a)/15d-14(a) Certification of Principal Financial Officer

 

X

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

32*

 

Section 1350 Certifications of Principal Executive Officer and Principal Financial Officer

 

X

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

101.INS

 

Inline XBRL Instance Document – The instance document does not appear in the interactive data file because its Inline XBRL tags are embedded within the Inline XBRL document.

 

X

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

101.SCH

 

Inline XBRL Taxonomy Extension Schema Document

 

X

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

101.CAL

 

Inline XBRL Taxonomy Extension Calculation Linkbase Document

 

X

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

101.DEF

 

Inline XBRL Taxonomy Extension Definition Linkbase Document

 

X

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

101.LAB

 

Inline XBRL Taxonomy Extension Label Linkbase Document

 

X

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

101.PRE

 

Inline XBRL Taxonomy Extension Presentation Linkbase Document

 

X

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

104

 

Cover Page Interactive Data File (formatted in Inline XBRL and contained in Exhibit 101)

 

X

 

 

 

 

 

 

 

 

(*) The certifications attached as Exhibit 32 that accompany this Quarterly Report on Form 10-Q are not deemed filed with the Securities and Exchange Commission and are not to be incorporated by reference into any filing of Wave Life Sciences Ltd. under the Securities Act of 1933, as amended, or the Securities Exchange Act of 1934, as amended (whether made before or after the date of such Form 10-Q), irrespective of any general incorporation language contained in such filing.

35


 

SIGNATURES

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

 

 

 

WAVE LIFE SCIENCES LTD.

 

 

 

 

Date: August 11, 2022

By:

 

/s/ Paul B. Bolno, M.D., MBA

 

 

 

Paul B. Bolno, M.D., MBA

 

 

 

President and Chief Executive Officer

 

 

 

(Principal Executive Officer)

 

 

 

 

Date: August 11, 2022

By:

 

/s/ Kyle Moran

 

 

 

Kyle Moran

 

 

 

Chief Financial Officer (Principal Financial Officer and Principal Accounting Officer)

 

36


EX-10.1

 

Exhibit 10.1

 

733 CONCORD AVENUE

CAMBRIDGE, MASSACHUSETTS

(the “Building”)

SECOND AMENDMENT

(“Second Amendment”)

 

EXECUTION DATE:

August 8, 2022

LANDLORD:

CPI/King 733 Concord Owner, LLC, a Delaware limited liability company (as successor-in-interest to King 733 Concord LLC, a Delaware limited liability company)

TENANT:

Wave Life Sciences USA, Inc., a Delaware corporation

EXISTING
PREMISES:

Approximately 44,019 rentable square feet of space, in the aggregate, consisting of: (i) approximately 30,893 rentable square feet of space in the Building (the “Original Premises”), and (ii) approximately 13,126 rentable square feet of space on the first (1st) floor of the Building (the “ROFO Premises”), both, as more particularly described in the Existing Lease (as defined below).

DATE OF EXISTING LEASE:

April 6, 2015

CURRENT EXPIRATION DATE OF ORIGINAL PREMISES:

EXTENDED EXPIRATION DATE FOR ORIGINAL PREMISES:

EXPIRATION DATE FOR ROFO PREMISES:

PRIOR AMENDMENTS/SUPPLEMENTS TO LEASE:

March 31, 2023

 

March 31, 2028

 

September 30, 2026

 

Commencement Letter dated October 1, 2015
Letter Agreement dated September 28, 2016
First Amendment dated December 9, 2020 (“
First Amendment”)
Term Commencement Letter dated September 30, 2021

WHEREAS, Tenant desires to extend the Term of the Lease with respect to the Original Premises for an additional period; and

WHEREAS, Landlord is willing to extend the Term of the Lease with respect to the Original Premises for an additional period, upon the terms and conditions hereinafter set forth.

NOW, THEREFORE, the Existing Lease, as previously amended, is hereby further amended by this Second Amendment (the Existing Lease, as previously amended, and as further amended by this Second Amendment, shall hereafter be referred to as the “Lease”). Any capitalized terms used herein shall have the same definition as set forth in the Existing Lease, except to the extent otherwise set forth in this Second Amendment.

1

 

 


 

1.
Extension of Term of Lease With Respect to the Original Premises. The Term of the Lease with respect to the Original Premises is hereby extended for an additional period (“Original Premises Extension Term”) commencing as of April 1, 2023 (the “Original Premises Extension Date”) and expiring as of March 31, 2028 (the “Original Premises Extended Expiration Date”), the parties hereby acknowledging and agreeing that the Expiration Date with respect to the ROFO Premises is September 30, 2026. The Original Premises Extension Term shall be on all of the terms and conditions of the Lease immediately preceding the Original Premises Extension Term, except as expressly set forth in this Second Amendment.
2.
Condition of Original Premises. Tenant hereby accepts the Original Premises in their “as is”, “where is” condition on the Original Premises Extension Date, with all faults, and without representations or warranties from Landlord of any kind whatsoever. Except for Landlord’s Original Premises Contribution (as hereinafter defined) and Landlord’s continuing maintenance and repair obligations expressly set forth in the Lease, Tenant acknowledges and agrees that Landlord has no obligation to perform any work or to provide any other concession with respect to the Original Premises for the Original Premises Extension Term.
3.
Base Rent. From and after the Original Premises Extension Date and continuing through the Original Premises Extended Expiration Date, Tenant shall pay Base Rent with respect to the Original Premises as follows:

 

Period of Time

 

Annual Base Rent

 

Monthly Base Rent

4/1/2023-3/31/2024:

$3,151,086.00

$262,590.50

4/1/2024-3/31/2025:

$3,245,618.58

$270,468.20

4/1/2025-3/31/2026:

$3,342,987.14

$278,582.25

4/1/2026-3/31/2027:

$3,443,276.75

$286,939.72

4/1/2027-3/31/2028:

$3,546,575.05

$295,547.91

 

4.
Additional Rent. During the Original Premises Extension Term, Tenant shall continue to pay to Landlord with respect to the Original Premises (i) Tenant’s Share of Operating Costs in accordance with the provisions of Section 5.2(c) of the Lease, (ii) Tenant’s Share of Taxes in accordance with the provisions of Section 5.3(c) of the Lease and (iii) the cost of all utility services consumed in the Original Premises (including, without limitation, electricity, gas and water) in accordance with the provisions of Section 9 of the Lease.
5.
Landlord’s Original Premises Contribution.
A.
Amount. Landlord shall provide to Tenant a tenant improvement allowance with respect to the Original Premises equal to up to Nine Hundred Twenty-Six Thousand Seven Hundred Ninety and 00/100 Dollars ($926,790.00) (“Landlord’s Original Premises Contribution”) to be used by Tenant solely for costs incurred by Tenant for work (“Tenant’s Work”) to be performed by Tenant in the Original Premises and/or the ROFO Premises. For the purposes hereof, Landlord’s Original Premises Contribution shall not include: (i) the cost of any of Tenant’s Property (as defined in Section 14.1(b) of the Lease), including without limitation telecommunications and computer equipment and all associated wiring and cabling and trade fixtures, (ii) the cost of any fixtures or Alterations (as defined in Section 11 of the Lease) that will be removed at the end of the Term, (iii) any fees paid to Tenant, any Affiliated Entity or Successor (as both terms are defined in Section 13.7 of the Lease), and (iv) any so-called “soft costs.” Tenant’s Work shall be performed in accordance with the provisions of the Lease (including, without limitation, Section 11 thereof).

2

 

 


 

B.
Requisitions. Landlord shall pay Landlord’s Proportion (hereinafter defined) of the cost shown on each requisition (hereinafter defined) submitted by Tenant to Landlord within thirty (30) days of submission thereof by Tenant to Landlord until the entirety of Landlord’s Original Premises Contribution has been exhausted. “Landlord’s Proportion” shall be a fraction, the numerator of which is Landlord’s Original Premises Contribution and the denominator of which is the total contract price for Tenant’s Work for the Original Premises (as evidenced by reasonably detailed documentation delivered to Landlord with the requisition first submitted by Tenant). A “requisition” shall mean AIA Documents G-702 and G-703 duly executed and certified by Tenant’s architect and general contractor (accompanied by, without limitation, invoices from Tenant’s contractors, vendors, service providers and consultants (collectively, “Contractors”) and partial lien waivers and subordinations of lien, as specified in M.G.L. Chapter 254, Section 32 (“Lien Waivers”) with respect to the prior month’s requisition, and such other documentation as Landlord or any Mortgagee may reasonably request) showing in reasonable detail the costs of the item in question or of the improvements installed to date in the Original Premises, accompanied by certifications executed by an officer of Tenant that the amount of the requisition in question does not exceed the cost of the items, services and work covered by such requisition. Landlord shall have the right, upon reasonable advance notice to Tenant, to inspect Tenant’s books and records relating to each requisition in order to verify the amount thereof. Tenant shall submit requisition(s) no more often than monthly. Landlord shall have no obligation to pay any portion of Landlord’s Original Premises Contribution with respect to any requisition submitted after April 1, 2025. Tenant shall not be entitled to any unused portion of Landlord’s Original Premises Contribution. Landlord’s obligation to pay any portion of Landlord’s Original Premises Contribution shall be conditioned upon there existing no default by Tenant in its obligations under the Lease which is continuing beyond any applicable notice and cure period at the time that Landlord would otherwise be required to make such payment. Landlord’s obligation to pay the final ten percent (10%) of Landlord’s Original Premises Contribution shall be subject to simultaneous delivery of all Lien Waivers relating to items, services and work performed in connection with Tenant’s Work.
6.
Notice Addresses. For all purposes of the Lease, the suite number for notice addresses for Landlord as set forth in Section 24 of the Lease, as amended by Section 3 of the First Amendment, has been changed to Suite 2400.
7.
Letter of Credit. The parties hereby acknowledge that Landlord is currently holding a Letter of Credit in the amount of $1,000,000.00 as the Security Amount pursuant to Section 7 of the Lease. The parties hereby further acknowledge that Landlord shall continue to hold said Letter of Credit during the Extension Term pursuant to and in accordance with said Section 7 of the Lease, except that the provisions of Section 7.6 of the Lease shall have no further applicability with respect to the Security Amount.
8.
Miscellaneous.
A.
Notice of Lease. In accordance with Section 25.16 of the Existing Lease, at Tenant’s option, each of the parties shall join in the execution, in recordable form and substantially similar to the form attached as Exhibit 10 to the Existing Lease, of a statutory notice of lease and/or written declaration, or an amendment to any existing notice and/or declaration, reflecting Tenant’s extension of the Term of the Lease, which notice of lease may be recorded by Tenant with the Middlesex South Registry of Deeds and/or filed with the Registry District of the Land Court, as appropriate.
B.
Broker. Tenant and Landlord each warrants and represents that it has dealt with no broker in connection with the consummation of this Second Amendment, other than Colliers International (the “Broker”). Tenant and Landlord each agrees to defend, indemnify and save the other harmless from and against any claims arising in breach of the representation and warranty set forth in the immediately preceding sentence. Landlord shall pay a brokerage commission to the Broker pursuant to a separate agreement between Landlord and the Broker.
C.
Deleted/Inapplicable Lease Provisions. Section 1.2 of the Lease (Extension Term) is hereby deleted in its entirety and is of no further force or effect. Section 3 and Exhibit 3 of the Lease shall have no applicability with respect to this Second Amendment.

3

 

 


 

D.
Ratification. In all other respects, except as expressly modified herein, the Lease is hereby ratified and confirmed. The submission of drafts of this document for examination and negotiation does not constitute an offer, or a reservation of or option for any of the terms and conditions set forth in this Second Amendment, and this Second Amendment shall not be binding upon Landlord or Tenant unless and until Landlord shall have executed and delivered a fully executed copy of this Second Amendment to Tenant.
E.
Conflict. In the event that any of the provisions of the Existing Lease are inconsistent with this Second Amendment or the state of facts contemplated hereby, the provisions of this Second Amendment shall control.
F.
Counterparts. This Second Amendment may be executed in any number of counterparts and by each of the undersigned on separate counterparts, which counterparts taken together shall constitute one and the same instrument. This Second Amendment may be executed by electronic signature, which shall be considered as an original signature for all purposes and shall have the same force and effect as an original signature. Without limitation, in addition to electronically produced signatures, “electronic signature” shall include faxed versions of an original signature or electronically scanned and transmitted versions (e.g., via pdf) of an original signature.

[Signature Page Follows]

4

 

 


 

EXECUTED as of the date first above written.

LANDLORD:

CPI/KING 733 CONCORD OWNER, LLC,

a Delaware limited liability company

 

By: /s/ Thomas Ragno__________________

Name: Thomas Ragno________________

Title: Authorized Signatory__________

 

 

TENANT:

WAVE LIFE SCIENCES USA, INC.
a Delaware corporation

 

By: /s/ Kyle Moran_____________________

Name: Kyle Moran___________________

Title: Chief Financial Officer___________

 

5

 

 


EX-31.1

 

Exhibit 31.1

CERTIFICATIONS UNDER SECTION 302

I, Paul B. Bolno, M.D., MBA, certify that:

 

1.
I have reviewed this Quarterly Report on Form 10-Q of Wave Life Sciences Ltd.;
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(s) 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 control 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(s) 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.

Dated: August 11, 2022

 

By:

 

/s/ Paul B. Bolno, M.D., MBA

 

 

Paul B. Bolno, M.D., MBA

 

 

President and Chief Executive Officer

 

 

(Principal Executive Officer)

 

 


EX-31.2

 

Exhibit 31.2

CERTIFICATIONS UNDER SECTION 302

I, Kyle Moran, certify that:

1.
I have reviewed this Quarterly Report on Form 10-Q of Wave Life Sciences Ltd.;
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(s) 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 control 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(s) 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.

Dated: August 11, 2022

 

By:

 

/s/ Kyle Moran

 

 

Kyle Moran

 

 

Chief Financial Officer

 

 

(Principal Financial Officer)

 

 

 


EX-32

 

Exhibit 32

CERTIFICATIONS UNDER SECTION 906

Pursuant to section 906 of the Sarbanes-Oxley Act of 2002 (subsections (a) and (b) of section 1350, chapter 63 of title 18, United States Code), each of the undersigned officers of Wave Life Sciences Ltd. (the “Company”), does hereby certify, to such officer’s knowledge, that:

The Quarterly Report for the quarter ended June 30, 2022 (the “Form 10-Q”) of the Company fully complies with the requirements of Section 13(a) or 15(d) of the Securities Exchange Act of 1934, and the information contained in the Form 10-Q fairly presents, in all material respects, the financial condition and results of operations of the Company.

 

Dated: August 11, 2022

 

/s/ Paul B. Bolno, M.D., MBA

 

 

Paul B. Bolno, M.D., MBA

 

 

President and Chief Executive Officer

 

 

(Principal Executive Officer)

 

Dated: August 11, 2022

 

/s/ Kyle Moran

 

 

Kyle Moran

 

 

Chief Financial Officer

 

 

(Principal Financial Officer)