8-K
SG false 0001631574 0001631574 2021-03-04 2021-03-04

 

 

UNITED STATES

SECURITIES AND EXCHANGE COMMISSION

Washington, D.C. 20549

 

 

Form 8-K

 

 

CURRENT REPORT

Pursuant to Section 13 or 15(d)

of the Securities Exchange Act of 1934

Date of Report (Date of earliest event reported): March 4, 2021

 

 

WAVE LIFE SCIENCES LTD.

(Exact name of registrant as specified in its charter)

 

 

 

Singapore   001-37627   00-0000000
(State or other jurisdiction
of incorporation)
 

(Commission

File Number)

 

(IRS Employer

Identification No.)

 

7 Straits View #12-00, Marina One

East Tower

Singapore

  018936
(Address of principal executive offices)   (Zip Code)

Registrant’s telephone number, including area code: +65 6236 3388

 

 

Check the appropriate box below if the Form 8-K filing is intended to simultaneously satisfy the filing obligation of the registrant under any of the following provisions (see General Instruction A.2. below):

 

Written communications pursuant to Rule 425 under the Securities Act (17 CFR 230.425)

 

Soliciting material pursuant to Rule 14a-12 under the Exchange Act (17 CFR 240.14a-12)

 

Pre-commencement communications pursuant to Rule 14d-2(b) under the Exchange Act (17 CFR 240.14d-2(b))

 

Pre-commencement communications pursuant to Rule 13e-4(c) under the Exchange Act (17 CFR 240.13e-4(c))

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 is an emerging growth company as defined in Rule 405 of the Securities Act of 1933 (§230.405 of this chapter) or Rule 12b-2 of the Securities Exchange Act of 1934 (§240.12b-2 of this chapter).

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.  ☐

 

 

 


Item 2.02

Results of Operations and Financial Condition.

On March 4, 2021, Wave Life Sciences Ltd. (the “Company”) announced its financial results for the quarter and year ended December 31, 2020. The full text of the press release issued in connection with the announcement is furnished as Exhibit 99.1 to this Current Report on Form 8-K and is incorporated by reference herein.

 

Item 7.01

Regulation FD Disclosure.

From time to time, the Company presents and/or distributes slides and presentations to the investment community to provide updates and summaries of its business. On March 4, 2021, the Company updated its corporate presentation, which is available on the “For Investors & Media” section of the Company’s website at http://ir.wavelifesciences.com/. This presentation is also furnished as Exhibit 99.2 to this Current Report on Form 8-K.

The information in these Items 2.02 and 7.01 are being furnished and shall not be deemed “filed” for purposes of Section 18 of the Securities Exchange Act of 1934, as amended (the “Exchange Act”), or otherwise subject to the liabilities of that Section, nor shall they be deemed incorporated by reference into any registration statement or other filing under the Securities Act of 1933, as amended, or the Exchange Act, except as shall be expressly set forth by specific reference in such filing.

 

Item 9.01

Financial Statements and Exhibits.

 

(d)

Exhibits

The following exhibits relating to Items 2.02 and 7.01 are furnished and not filed:

 

Exhibit
No.
   Description
99.1    Press Release issued by Wave Life Sciences Ltd. dated March 4, 2021
99.2    Corporate Presentation of Wave Life Sciences Ltd. dated March 4, 2021
104    Cover Page Interactive Data File (embedded within the Inline XBRL document)


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 hereunto duly authorized.

 

WAVE LIFE SCIENCES LTD.
By:  

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

  Paul B. Bolno, M.D.
  President and Chief Executive Officer

Date: March 4, 2021

EX-99.1

Exhibit 99.1

 

LOGO

Wave Life Sciences Reports Fourth Quarter and Full Year 2020 Financial Results and Provides Business Update

Strong execution in 2020 sets stage for advancing five clinical programs and novel ADAR editing modality in 2021

Data from ongoing PRECISION-HD and OLE clinical trials for Huntington’s disease on track for end of 1Q 2021

Moving towards first patient dosing in three clinical trials with pipeline candidates incorporating PN chemistry: WVE-003

(SNP3), WVE-004 (C9orf72) and WVE-N531 (Exon 53)

Cash runway expected into 2Q 2023

Wave to host investor conference call and webcast at 8:00 a.m. ET today

CAMBRIDGE, Mass., Mar. 4, 2021 (GLOBE NEWSWIRE) — Wave Life Sciences Ltd. (Nasdaq: WVE), a clinical-stage genetic medicines company committed to delivering life-changing treatments for people battling devastating diseases, today announced financial results for the fourth quarter and full year ended December 31, 2020 and provided a business update.

“Wave is entering 2021 with depth and diversity throughout our pipeline and platform, the result of focused and deliberate execution, and a steadfast commitment to leading a new era of RNA therapeutics. Despite headwinds from the COVID-19 pandemic, we advanced our pipeline, significantly evolved our platform, announced our first ADAR editing program and added considerable talent to our innovative and driven team,” said Paul Bolno, MD, MBA, President and Chief Executive Officer of Wave Life Sciences. “We are poised to bring five clinical programs forward in 2021, including three programs in Huntington’s disease, a fourth program for ALS and FTD and a fifth program for exon 53 skipping in DMD. We remain on track to announce data from the Phase 1b/2a PRECISION-HD1 and PRECISION-HD2 trials at the end of the first quarter of 2021 and are excited about adding our next set of clinical programs incorporating novel PN backbone chemistry modifications, which preclinically have been shown to increase potency, exposure and durability in our growing portfolio of investigational stereopure oligonucleotides. Finally, we strengthened our balance sheet in September 2020 to support our pipeline and discovery work, extending our cash runway into the second quarter of 2023 and ensuring Wave is well-positioned to unlock potential and growth well beyond 2021.”

2020 Full Year and Recent Business Highlights and Upcoming Milestones

Three programs with novel PN backbone chemistry modifications expected to enter clinic in 2021:

 

   

In August 2020, Wave introduced novel PN backbone chemistry modifications, an advancement from its PRISMTM discovery and drug development platform. In preclinical studies, these modifications have been shown to increase potency, exposure and durability across silencing, splicing and RNA editing modalities.

 

   

Wave expects to initiate dosing in three clinical trials in 2021, which will assess target engagement, impact on key disease biomarkers, and initial safety of WVE-003 (targeting SNP3), WVE-004 (targeting C9orf72) and WVE-N531 (targeting exon 53).


   

All three compounds were designed with PN backbone chemistry modifications, and insight from pharmacokinetic (PK) and pharmacodynamic (PD) studies using in vivo models, as well as learnings from Wave’s first-generation programs.

Programs for Huntington’s disease (HD): Wave is developing a unique portfolio of investigational stereopure oligonucleotides designed to selectively target the mutant allele of the huntingtin (mHTT) gene, while leaving the wild-type (wtHTT) protein relatively intact. Wave’s approach to HD is guided by the recognition that, in addition to a gain of function of the mHTT protein, people with this disease have lost one copy of the wtHTT allele, leaving them with a smaller protective reservoir of healthy protein than unaffected individuals. A growing body of scientific evidence suggests that preserving as much of this essential protein as possible is important for favorable health outcomes. Wave’s allele-selective approach may also enable treatment in the premanifest setting, before onset of clinical disease.

PRECISION-HD and OLE clinical trials in HD (WVE-120101 and WVE-120102):

 

   

The PRECISION-HD1 and PRECISION-HD2 Phase 1b/2a trials evaluating investigational WVE-120101 (SNP1) and WVE-120102 (SNP2), respectively, in patients with HD are ongoing. WVE-120101 and WVE-120102 are designed to selectively target the mHTT mRNA transcript that contains specific single nucleotide polymorphisms (SNPs).

 

   

Open-label extension (OLE) clinical trials for patients outside of the U.S. who participated in the Phase 1b/2a PRECISION-HD trials are also ongoing.

 

   

Wave expects to report biomarker and safety data from all cohorts of the PRECISION-HD2 trial, along with data from all completed cohorts up to and including the 16 mg cohort from the PRECISION-HD1 trial at the end of the first quarter of 2021. Wave also expects to report data from patients who have received multiple doses of 8 or 16 mg of WVE-120101 or WVE-120102 in the OLE trials at the end of the first quarter of 2021.

WVE-003 (SNP3) for HD:

 

   

WVE-003 is Wave’s first allele-selective HD candidate that uses PN backbone chemistry modifications and was developed using preclinical in vivo models to enable target engagement assessment of a specific single nucleotide polymorphism (SNP3). In preclinical studies, WVE-003 showed selective reduction of mHTT mRNA in vitro, and potent and durable knockdown of mHTT mRNA in vivo.

 

   

In December 2020, Wave initiated clinical development of WVE-003 with the submission of a clinical trial application (CTA).

 

   

Wave expects to initiate dosing in a Phase 1b/2a clinical trial of WVE-003 for patients with HD in 2021.

Publications:

 

   

In December 2020, in Molecular Therapy Methods & Clinical Development, Wave published its haplotype phasing method using single-molecule real-time sequencing and a custom algorithm to determine bases at SNPs on mutant alleles. Accurate haplotype phasing of SNPs and the expanded CAG repeat of the huntingtin gene enables identification of patients with Huntington’s disease eligible for allele-selective clinical studies.

 

   

In May 2020, Wave’s prospective observational study of the frequency of SNP1 and SNP2 in patients with HD was published in Neurology Genetics. The study confirms the feasibility of rapidly and prospectively identifying SNP1 and/or SNP2 in association with the mHTT allele in patients with HD, to enable allele-selective, personalized treatment approaches in eligible patients.

WVE-004 (C9orf72) for amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD):

 

   

In February 2021, Wave published in Nature Communications results of initial work to identify and validate its targeting strategy to achieve variant-selective knockdown of expansion-containing C9orf72 transcripts with stereopure oligonucleotides for the treatment of ALS and FTD. The results in the publication represent the foundational work that led to the development of Wave’s clinical candidate, WVE-004, which uses PN backbone chemistry modifications.

 

   

In December 2020, Wave initiated clinical development of WVE-004 with the submission of a CTA.


   

In August 2020, Wave presented preclinical in vivo data for WVE-004, which demonstrated potent and durable knockdown of more than 90% of poly GP dipeptide repeat (DPR) protein in the spinal cord and at least 80% in the cortex, an effect that persisted for at least six months. Healthy C9orf72 protein was relatively unchanged over the same time period.

 

   

Wave expects to initiate dosing in a Phase 1b/2a clinical trial of WVE-004 for both patients with C9-ALS and patients with C9-FTD in 2021.

WVE-N531 for Duchenne muscular dystrophy (DMD) amenable to exon 53 skipping:

 

   

WVE-N531 is Wave’s first splicing candidate to incorporate PN backbone chemistry modifications.

 

   

In a recently completed in vivo study of double knock-out mice (a model lacking dystrophin and utrophin protein with a severe phenotype), an oligonucleotide designed with PN backbone chemistry modifications appeared to significantly increase dystrophin production and substantially improve survival.

 

   

In a planned clinical trial, Wave will assess dystrophin production and initial safety in patients with DMD amenable to exon 53 skipping.

 

   

Wave expects to submit a CTA for WVE-N531 by the end of the first quarter of 2021.

Central nervous system (CNS) programs in collaboration with Takeda:

 

   

Wave is utilizing PN backbone chemistry modifications to design stereopure oligonucleotides for CNS indications, including Alzheimer’s disease, Parkinson’s disease and others, as part of its ongoing collaboration with Takeda. Wave continues to produce compelling in vivo data and progress multiple discovery programs towards portfolio entry and candidate nomination.

 

   

In the fourth quarter of 2020, Wave achieved the first demonstration of widespread target engagement in the CNS of non-human primates (NHPs) for the most advanced therapeutic program in the collaboration. Approximately 90% knockdown of the target mRNA was observed one month after a single 12 mg intrathecal dose, and the therapeutic candidate distributed widely across relevant CNS tissues.

Alpha-1 antitrypsin deficiency (AATD) program with ADAR editing:

 

   

Wave’s AATD program, its first ADAR editing program, will target the G-to-A disease-causing mutation in mRNA coded by the SERPINA1 Z allele. By correcting the single RNA base mutation, ADAR editing may provide an ideal approach for increasing circulating levels of wild-type alpha-1 antitrypsin (AAT) protein and reducing aggregation in the liver, thus simultaneously addressing both the lung and liver manifestations of the disease.

 

   

In November 2020, Wave presented in vitro data in a primary hepatocyte SERPINA1 Z allele cell model, which demonstrated that editing the Z transcript back to wild-type prevents protein misfolding and increases secretion of edited AAT protein from hepatocytes.

 

   

Wave expects to deliver in vivo data supporting the continued development of its AATD program in the first half of 2021.

ADAR editing platform modality:

 

   

Wave’s novel RNA editing modality incorporates PN backbone chemistry modifications and uses endogenous ADAR (adenosine deaminases acting on RNA) enzymes via free uptake (non-viral, no nanoparticles) of A-to-I (G) RNA editing oligonucleotides. ADAR editing has the potential to unlock many new therapeutic applications, including restoration, modification or upregulation of proteins.

Fourth Quarter and Full Year 2020 Financial Results and Financial Guidance

Wave reported a net loss of $28.8 million in the fourth quarter of 2020 as compared to $56.8 million in the same period in 2019. The company reported a net loss of $149.9 million for the year ended December 31, 2020 as compared to $193.6 million for the year ended December 31, 2019.


Research and development expenses were $30.0 million in the fourth quarter of 2020 as compared to $49.1 million in the same period in 2019. Research and development expenses were $130.9 million in 2020, as compared to $175.4 million in 2019. The decrease in research and development expenses in the fourth quarter and full year was primarily due to the decrease in external expenses related to Wave’s decision to discontinue its suvodirsen program in December 2019, as well as decreases in compensation-related expenses and other external expenses driven by Wave’s February 2020 cost reduction plan, partially offset by the increases in external expenses related to Wave’s clinical and preclinical activities related to its HD programs and its C9orf72 program for ALS and FTD.

General and administrative expenses were $9.7 million in the fourth quarter of 2020, as compared to $13.8 million in the same period in 2019. General and administrative expenses were $42.5 million in 2020, as compared to $48.9 million in 2019. The decrease in general and administrative expenses in the fourth quarter and full year was primarily driven by the February 2020 cost reduction plan, which led to decreases in compensation-related expenses and other external expenses.

Wave ended 2020 with $184.5 million in cash and cash equivalents as compared to $147.2 million as of December 31, 2019. During 2020, Wave substantially extended its cash runway, largely by raising $93.7 million in net proceeds from its September 2020 public offering and $59.9 million in net proceeds from its at-the-market equity program.

Wave expects that its existing cash and cash equivalents, together with expected and committed cash from its existing collaboration, will enable the company to fund its operating and capital expenditure requirements into the second quarter of 2023.

Investor Conference Call and Webcast

Wave management will host an investor conference call today at 8:00 a.m. ET to discuss the company’s fourth quarter and full year 2020 financial results and provide a business update. The conference call may be accessed by dialing (866) 220-8068 (domestic) or (470) 495-9153 (international) and entering conference ID: 6269069. The live webcast may be accessed from the investor relations section of the Wave Life Sciences corporate website at ir.wavelifesciences.com. Following the webcast, a replay will be available on the website.

About PRISM

PRISM is Wave Life Sciences’ proprietary discovery and drug development platform that enables genetically defined diseases to be targeted with stereopure oligonucleotides across multiple therapeutic modalities, including silencing, splicing and editing. PRISM combines the company’s unique ability to construct stereopure oligonucleotides with a deep understanding of how the interplay among oligonucleotide sequence, chemistry and backbone stereochemistry impacts key pharmacological properties. By exploring these interactions through iterative analysis of in vitro and in vivo outcomes and machine learning-driven predictive modeling, the company continues to define design principles that are deployed across programs to rapidly develop and manufacture clinical candidates that meet pre-defined product profiles.

About Wave Life Sciences

Wave Life Sciences (Nasdaq: WVE) is a clinical-stage genetic medicines company committed to delivering life-changing treatments for people battling devastating diseases. Wave aspires to develop best-in-class medicines across multiple therapeutic modalities using PRISM, the company’s proprietary discovery and drug development platform that enables the precise design, optimization and production of stereopure oligonucleotides. Driven by a resolute sense of urgency, the Wave team is targeting a broad range of genetically defined diseases so that patients and families may realize a brighter future. To find out more, please visit www.wavelifesciences.com and follow Wave on Twitter @WaveLifeSci.

Forward-Looking Statements

This press release contains forward-looking statements concerning our goals, beliefs, expectations, strategies, objectives and plans, and other statements that are not necessarily based on historical facts, including statements regarding the following, among others: the anticipated commencement, patient enrollment, data readouts and completion of our clinical trials, and the announcement of such events; the protocol, design and endpoints of our ongoing and planned


clinical trials; the future performance and results of our programs in clinical trials; future preclinical activities and programs; regulatory submissions; the progress and potential benefits of our collaborations with partners; the potential of our in vitro and in vivo preclinical data to predict the behavior of our compounds in humans; our identification of future product candidates and their therapeutic potential; the anticipated therapeutic benefits of our potential therapies compared to others; our ability to design compounds using multiple modalities and the anticipated benefits of that model; the anticipated benefits of our proprietary manufacturing processes and our internal manufacturing capabilities; the potential benefits of PRISM, including our novel PN backbone chemistry modifications, and our stereopure oligonucleotides compared with stereorandom oligonucleotides; the potential benefits of our novel ADAR-mediated RNA editing platform capabilities compared to others; the benefit of nucleic acid therapeutics generally; the strength of our intellectual property; the anticipated duration of our cash runway; and our expectations regarding the impact of the COVID-19 pandemic on our business. Actual results may differ materially from those indicated by these forward-looking statements as a result of various important factors, including the following: our ability to finance our drug discovery and development efforts and to raise additional capital when needed; the ability of our preclinical programs to produce data sufficient to support our clinical trial applications and the timing thereof; our ability to maintain the company infrastructure and personnel needed to achieve our goals; the clinical results of our programs, which may not support further development of product candidates; actions of regulatory agencies, which may affect the initiation, timing and progress of clinical trials; our effectiveness in managing future clinical trials and regulatory interactions; the effectiveness of PRISM, including our novel PN backbone chemistry modifications ; the effectiveness of our novel ADAR-mediated RNA editing platform capability; the continued development and acceptance of oligonucleotides as a class of medicines; our ability to demonstrate the therapeutic benefits of our candidates in clinical trials, including our ability to develop candidates across multiple therapeutic modalities; our dependence on third parties, including contract research organizations, contract manufacturing organizations, collaborators and partners; our ability to manufacture or contract with third parties to manufacture drug material to support our programs and growth; our ability to obtain, maintain and protect our intellectual property; our ability to enforce our patents against infringers and defend our patent portfolio against challenges from third parties; competition from others developing therapies for similar indications; the severity and duration of the COVID-19 pandemic and its negative impact on the conduct of, and the timing of enrollment, completion and reporting with respect to, our clinical trials; and any other impacts on our business as a result of or related to the COVID-19 pandemic, as well as the information under the caption “Risk Factors” contained in our most recent Annual Report on Form 10-K filed with the Securities and Exchange Commission (SEC) and in other filings we make with the SEC from time to time. We undertake no obligation to update the information contained in this press release to reflect subsequently occurring events or circumstances.


WAVE LIFE SCIENCES LTD.

UNAUDITED CONSOLIDATED BALANCE SHEETS

(In thousands, except share amounts)

 

     December 31, 2020     December 31, 2019  

Assets

    

Current assets:

    

Cash and cash equivalents

   $ 184,497     $ 147,161  

Current portion of accounts receivable

     30,000       20,000  

Prepaid expenses

     10,434       9,626  

Other current assets

     5,111       8,689  
  

 

 

   

 

 

 

Total current assets

     230,042       185,476  

Long-term assets:

    

Accounts receivable, net of current portion

     —         30,000  

Property and equipment, net

     29,198       36,368  

Operating lease right-of-use assets

     16,232       18,101  

Restricted cash

     3,651       3,647  

Other assets

     115       10,658  
  

 

 

   

 

 

 

Total long-term assets

     49,196       98,774  
  

 

 

   

 

 

 

Total assets

   $ 279,238     $ 284,250  
  

 

 

   

 

 

 

Liabilities, Series A preferred shares and shareholders’ equity

    

Current liabilities:

    

Accounts payable

   $ 13,795     $ 9,073  

Accrued expenses and other current liabilities

     11,971       16,185  

Current portion of deferred revenue

     91,560       89,652  

Current portion of operating lease liability

     3,714       3,243  
  

 

 

   

 

 

 

Total current liabilities

     121,040       118,153  

Long-term liabilities:

    

Deferred revenue, net of current portion

     41,481       63,466  

Operating lease liability, net of current portion

     25,591       29,304  

Other liabilities

     474       1,721  
  

 

 

   

 

 

 

Total long-term liabilities

     67,546       94,491  
  

 

 

   

 

 

 

Total liabilities

   $ 188,586     $ 212,644  
  

 

 

   

 

 

 

Series A preferred shares, no par value; 3,901,348 shares issued and outstanding at December 31, 2020 and 2019

   $ 7,874     $ 7,874  
  

 

 

   

 

 

 

Shareholders’ equity:

    

Ordinary shares, no par value; 48,778,678 and 34,340,690 shares issued and outstanding at December 31, 2020 and 2019, respectively

     694,085       539,547  

Additional paid-in capital

     71,573       57,277  

Accumulated other comprehensive income

     389       267  

Accumulated deficit

     (683,269     (533,359
  

 

 

   

 

 

 

Total shareholders’ equity

     82,778       63,732  
  

 

 

   

 

 

 

Total liabilities, Series A preferred shares and shareholders’ equity

   $ 279,238     $ 284,250  
  

 

 

   

 

 

 


WAVE LIFE SCIENCES LTD.

UNAUDITED CONSOLIDATED STATEMENTS OF OPERATIONS AND COMPREHENSIVE LOSS

(In thousands, except share and per share amounts)

 

     Three Months Ended December 31,     Twelve Months Ended December 31,  
     2020     2019     2020     2019  

Revenue

   $ 9,439     $ 2,400     $ 20,077     $ 15,983  
  

 

 

   

 

 

   

 

 

   

 

 

 

Operating expenses:

        

Research and development

     30,033       49,128       130,944       175,431  

General and administrative

     9,719       13,805       42,510       48,869  
  

 

 

   

 

 

   

 

 

   

 

 

 

Total operating expenses

     39,752       62,933       173,454       224,300  
  

 

 

   

 

 

   

 

 

   

 

 

 

Loss from operations

     (30,313     (60,533     (153,377     (208,317

Other income, net:

        

Dividend income

     24       736       584       4,912  

Interest income (expense), net

     —         4       (16     29  

Other income, net

     659       3,023       2,058       9,738  
  

 

 

   

 

 

   

 

 

   

 

 

 

Total other income, net

     683       3,763       2,626       14,679  
  

 

 

   

 

 

   

 

 

   

 

 

 

Loss before income taxes

     (29,630     (56,770     (150,751     (193,638

Income tax benefit (provision), net

     841       —         841       —    
  

 

 

   

 

 

   

 

 

   

 

 

 

Net loss

   $ (28,789   $ (56,770   $ (149,910   $ (193,638
  

 

 

   

 

 

   

 

 

   

 

 

 

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

   $ (0.59   $ (1.65   $ (3.82   $ (5.72
  

 

 

   

 

 

   

 

 

   

 

 

 

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

     48,777,001       34,303,975       39,227,618       33,866,487  
  

 

 

   

 

 

   

 

 

   

 

 

 

Other comprehensive income (loss):

        

Net loss

   $ (28,789   $ (56,770   $ (149,910   $ (193,638

Foreign currency translation

     88       (15     122       114  
  

 

 

   

 

 

   

 

 

   

 

 

 

Comprehensive loss

   $ (28,701   $ (56,785   $ (149,788   $ (193,524
  

 

 

   

 

 

   

 

 

   

 

 

 

Investor Contact:

Kate Rausch

617-949-4827

krausch@wavelifesci.com

Media Contact:

Alicia Suter

617-949-4817

asuter@wavelifesci.com

EX-99.2

Slide 1

Wave Life Sciences Corporate Presentation March 4, 2021 Exhibit 99.2


Slide 2

Forward-looking statements This document contains forward-looking statements. All statements other than statements of historical facts contained in this document, including statements regarding possible or assumed future results of operations, preclinical and clinical studies, business strategies, research and development plans, collaborations and partnerships, regulatory activities and timing thereof, competitive position, potential growth opportunities, use of proceeds and the effects of competition are forward-looking statements. These statements involve known and unknown risks, uncertainties and other important factors that may cause the actual results, performance or achievements of Wave Life Sciences Ltd. (the “Company”) to be materially different from any future results, performance or achievements expressed or implied by the forward-looking statements. In some cases, you can identify forward-looking statements by terms such as “may,” “will,” “should,” “expect,” “plan,” “aim,” “anticipate,” “could,” “intend,” “target,” “project,” “contemplate,” “believe,” “estimate,” “predict,” “potential” or “continue” or the negative of these terms or other similar expressions. The forward-looking statements in this presentation are only predictions. The Company has based these forward-looking statements largely on its current expectations and projections about future events and financial trends that it believes may affect the Company’s business, financial condition and results of operations. These forward-looking statements speak only as of the date of this presentation and are subject to a number of risks, uncertainties and assumptions, including those listed under Risk Factors in the Company’s Form 10-K and other filings with the SEC, some of which cannot be predicted or quantified and some of which are beyond the Company’s control. The events and circumstances reflected in the Company’s forward-looking statements may not be achieved or occur, and actual results could differ materially from those projected in the forward-looking statements. Moreover, the Company operates in a dynamic industry and economy. New risk factors and uncertainties may emerge from time to time, and it is not possible for management to predict all risk factors and uncertainties that the Company may face. Except as required by applicable law, the Company does not plan to publicly update or revise any forward-looking statements contained herein, whether as a result of any new information, future events, changed circumstances or otherwise.


Slide 3

Building a leading genetic medicines company ALS: Amyotrophic lateral sclerosis; FTD: Frontotemporal dementia 1stereopure oligonucleotides and novel backbone chemistry modifications Innovative platform Stereopure oligonucleotides Novel backbone modifications (PN chemistry) Allele-selectivity Multiple modalities (silencing, splicing, ADAR editing) Strong IP position1 Foundation of NEUROLOGY programs Huntington’s disease ALS / FTD Neuromuscular diseases Ataxias Parkinson’s disease Alzheimer’s disease Clinical development expertise Multiple global clinical trials ongoing across eight countries Innovative trial designs Manufacturing Established internal manufacturing capabilities to produce oligonucleotides at scale Wave’s discovery and drug development platform


Slide 4

PRISM has unlocked novel and proprietary advances in oligonucleotide design Backbone modifications Sugar modifications Drug approvals (FDA)2 1975 2020 2000 Mixtures of 2n molecules1 ~500,000 different molecules per dose fomivirsen pegaptanib Phosphorothioate (PS) mipomersen nusinersen PN backbone chemistry modifications Stereopure backbone 2’-4’-cEt 2’-O-methyl 2’-F 2’-4’-LNA 1n=number of chiral centers 2’-MOE Phosphorodiamidate Morpholino (PMO) eteplirsen golodirsen givosiran patisiran inotersen viltolarsen 2oligonucleotide therapies approved by the FDA across the industry


Slide 5

THERAPEUTIC AREA / TARGET DISCOVERY PRECLINICAL CLINICAL PARTNER Huntington’s disease mHTT SNP1 Takeda 50:50 option Huntington’s disease mHTT SNP2 Huntington’s disease mHTT SNP3 ALS and FTD C9orf72 SCA3 ATXN3 CNS diseases Multiple† Takeda milestones & royalties DMD Exon 53 100% global ADAR editing Multiple AATD (ADAR editing) SERPINA1 100% global Retinal diseases USH2A and RhoP23H 100% global NEUROLOGY HEPATIC OPTHALMOLOGY WVE-003 WVE-004 WVE-120101 WVE-120102 Innovative pipeline led by neurology programs †During a four-year term, Wave and Takeda may collaborate on up to six preclinical targets at any one time. ALS: Amyotrophic lateral sclerosis; FTD: Frontotemporal dementia; SCA3: Spinocerebellar ataxia 3; CNS: Central nervous system; DMD: Duchenne muscular dystrophy; AATD: Alpha-1 antitrypsin deficiency Stereopure PN chemistry WVE-N531


Slide 6

Platform evolution reflected in three upcoming clinical trials to start in 2021 Oligonucleotide optimization Stereopure backbone PN backbone chemistry modifications In vivo disease models Insight into PK / PD relationships Novel model generation Leverage learnings of first generation programs Translational pharmacology Clinical trial design Allele-selective silencing candidate in HD WVE-003 SNP3 WVE-004 C9orf72 Variant-selective silencing candidate in ALS and FTD WVE-N531 Exon 53 Exon skipping candidate for DMD HD: Huntington’s diseaseALS: amyotrophic lateral sclerosisFTD: frontotemporal dementia DMD: Duchenne muscular dystrophy


Slide 7

WVE-120101 WVE-120102 WVE-003 Huntington’s Disease Portfolio


Slide 8

Huntington’s disease: a hereditary, fatal disorder Sources: Auerbach W, et al. Hum Mol Genet. 2001;10:2515-2523. Dragatsis I, et al. Nat Genet. 2000;26:300-306. Leavitt BR, et al. J Neurochem. 2006;96:1121-1129. Nasir J, et al. Cell. 1995;81:811-823. Reiner A, et al. J Neurosci. 2001;21:7608-7619. White JK, et al. Nat Genet. 1997;17:404-410. Zeitlin S, et al. Nat Genet. 1995;11:155-163. Carroll JB, et al. Mol Ther. 2011;19:2178-2185. HDSA ‘What is Huntington’s disease?’ https://hdsa.org/what-is-hd/overview-of-huntingtons-disease/ Accessed: 3/3/21. HDSA ‘Who is at Risk’ https://hdsa.org/what-is-hd/history-and-genetics-of-huntingtons-disease/who-is-at-risk/ Accessed: 3/3/21. Becanovic, K., et al., Nat Neurosci, 2015. 18(6): p. 807-16. Van Raamsdonk, J.M., et al., Hum Mol Genet, 2005. 14(10): p. 1379-92.; Van Raamsdonk, J.M., et al., BMC Neurosci, 2006. 7: p. 80. DNA CAG Repeat RNA wild-type (healthy) allele RNA mutant allele Normal CAG Repeat Expanded CAG Repeat Healthy protein (HTT) Mutant protein (mHTT) Neuro HD Autosomal dominant disease, characterized by cognitive decline, psychiatric illness and chorea; fatal No approved disease-modifying therapies Expanded CAG triplet repeat in HTT gene results in production of mutant huntingtin protein (mHTT); accumulation of mHTT causes progressive loss of neurons in the brain Wild-type (healthy) HTT protein critical for neuronal function; evidence suggests wild-type HTT loss of function plays a role in Huntington’s disease (HD) 30,000 people with HD in the US and more than 200,000 at risk of developing HD


Slide 9

Healthy CNS function Synaptic dysfunction | Cell death | Neurodegeneration mHTT toxic effects lead to neurodegeneration, loss of wtHTT functions may also contribute to HD Healthy individual Stresses wtHTT Huntington’s disease Stresses Toxic effects of mHTT + Loss of wtHTT functions ~50% decrease in wtHTT CNS, central nervous system; HD, Huntington’s disease; HTT, huntingtin protein; mHTT, mutant huntingtin protein; wtHTT, wild-type huntingtin protein. 1. Ross CA, Tabrizi SJ. Lancet Neurol. 2011;10(1):83-98. 2. Saudou F, Humbert S. Neuron. 2016;89(5):910-926. 3. Cattaneo E, et al. Nat Rev Neurosci. 2005;6(12):919-930. 4. Milnerwood AJ, Raymond LA. Trends Neurosci. 2010;33(11):513-523. Neuro HD


Slide 10

Plays an essential role in the transport of synaptic proteins—including neurotransmitters and receptors—to their correct location at synapses9-12 Promotes neuronal survival by protecting against stress (e.g., excitotoxicity, oxidative stress, toxic mHTT aggregates)1-8 BRAIN CIRCUITS SYNAPSE NEURON CSF circulation Supplies BDNF to the striatum to ensure neuronal survival13-16 Regulates synaptic plasticity, which underlies learning and memory17-22 Plays a critical role in formation and function of cilia—sensory organelles that control the flow of CSF—which are needed to clear catabolites and maintain homeostasis23 HD: Wild-type HTT is a critical protein for important functions in the central nervous system BDNF, brain-derived neurotrophic factor; CSF, cerebrospinal fluid; mHTT, mutant huntingtin protein. Sources: 1. Leavitt 2006 2. Cattaneo 2005 3. Kumar 2016 4. Franco-Iborra 2020 5. Hamilton 2015 6. Ochaba 2014 7. Wong 2014 8. Rui 2015 9. Caviston 2007 10. Twelvetrees 2010 11. Strehlow 2007 12. Milnerwood 2010 13. Smith-Dijak 2019 14. Tousley 2019 15. Zhang 2018 16. McAdam 2020 17. Altar 1997 18. Zuccato 2001 19. Gauthier 2004 20. Ferrer 2000 21. Baquet 2004 22. Liu 2011 23. Karam 2015 Neuro HD


Slide 11

Nature publication contributes to weight of evidence on importance of wild-type huntingtin Source: Poplawski et al., Nature, April 2019 Htt: Huntingtin protein Conditional knock-out of Htt in 4-month old mice (post-neuronal development) Results suggest that: Htt plays a central role in the regenerating transcriptome (potentially influencing genes such as NFKB, STAT3, BDNF) Htt is essential for regeneration Indeed, conditional gene deletion showed that Htt is required for neuronal repair. Throughout life, neuronal maintenance and repair are essential to support adequate cellular functioning Neuro HD


Slide 12

Utilize association between single nucleotide polymorphisms (SNPs) and genetic mutations to specifically target errors in genetic disorders, including Huntington’s disease (HD) Potential to provide treatment for up to 80% of HD population Wave approach: novel, allele-selective silencing Source: Kay, et al. Personalized gene silencing therapeutics for Huntington disease. Clin Genet. 2014;86:29–36. Neuro HD Aims to lower mHTT transcript while leaving healthy wild-type HTT relatively intact Allele-selectivity possible by targeting SNPs associated with expanded long CAG repeat in HTT gene RNase H and ASO:RNA RNA mutant allele


Slide 13

WVE-120101: Selective reduction of mHTT mRNA and protein Reporter Cell Line* Neuro HD Source: Meena, Zboray L, Svrzikapa N, et al. Selectivity and biodistribution of WVE-120101, a potential antisense oligonucleotide therapy for the treatment of Huntington’s disease. Paper presented at: 69th Annual Meeting of the American Academy of Neurology; April 28, 2017; Boston, MA.


Slide 14

Demonstrated delivery to brain tissue WVE-120101 and WVE-120102 distribution in cynomolgus non-human primate brain following intrathecal bolus injection In Situ Hybridization ViewRNA stained tissue Red dots are WVE-120102 oligonucleotide Arrow points to nuclear and perinuclear distribution of WVE-120102 in caudate nucleus Red dots are WVE-120101 oligonucleotide Arrow points to nuclear and perinuclear distribution of WVE- 120101 in cingulate cortex CIC = cingulate cortex In Situ Hybridization ViewRNA stained tissue  Neuro HD CN = caudate nucleus Source: Meena, Zboray L, Svrzikapa N, et al. Selectivity and biodistribution of WVE-120101, a potential antisense oligonucleotide therapy for the treatment of Huntington’s disease. Paper presented at: 69th Annual Meeting of the American Academy of Neurology; April 28, 2017; Boston, MA.


Slide 15

PRECISION-HD clinical trials Single Dose Multidose 196 1 Washout CSF sample Dose 28 56 84 112 Study Day* 140 OLE 2 mg 4 mg 8 mg 16 mg 32 mg Multidose Cohorts (N = 12 per cohort) OLE: Open label extension; CSF: cerebrospinal fluid; mHTT: mutant huntingtin; NfL: neurofilament light chain; wtHTT: wild-type HTT *Study day may vary depending on patient washout period Results Two Phase 1b/2a clinical trials for WVE-120101 and WVE-120102 ongoing Patients are migrated to highest dose tested Neuro HD Safety and tolerability Biomarkers mHTT wtHTT PRECISION-HD1 and OLE (including complete 16 mg cohort) PRECISION-HD2 and OLE (including complete 32 mg cohort) NfL Trial results expected end of 1Q 2021


Slide 16

Neuro HD Assessment of wild-type protein in CSF Ab: antibodyCSF: cerebrospinal fluidHTT: huntingtin proteinMW1: Wild et al. JCI 2015 CSF sample Total HTT mt wt wt wt mt mt mt mt mt wt wt wt Wild-type HTT wt wt wt wt wt wt wt wt Deplete mutant HTT Add polyQ Ab magnetic beads to CSF sample mt mt mt mt mt mt polyQ Ab magnetic beads Biotin polyQ Ab (MW1) Magnetic streptavidin beads Wild-type huntingtin protein Mutant huntingtin protein wt mt Depletion of mutant HTT key to ability to measure wild-type HTT protein


Slide 17

WVE-003 (SNP3) demonstrates selective, potent, and durable reduction of mHTT in preclinical models Selectively reduces mHTT mRNA in HD iPSC neurons in vitro Results from ND50036 iPSC-derived medium spiny neurons. Total HTT knockdown quantified by qPCR and normalized to HPRT1 Oligonucleotide or PBS [100 μg ICV injections through a cannula on days 1, 3, and 5] delivered to BACHD transgenic. Mean ± SD (n=8, *P<0.0332, ***P<0.0002, ****P<0.0001 versus PBS unless otherwise noted).  HPRT1, hypoxanthine-guanine phosphoribosyl transferase; iPSC, induced pluripotent stem cell; ICV, intracerebroventricular; PBS, phosphate-buffered saline Similar results in cortex Pan-silencing reference compound WVE-003 PBS Weeks *** **** **** **** **** **** Pan-silencing reference compound WVE-003 Percentage HTT mRNA Remaining Durable striatal mHTT knockdown for 12 weeks in  BACHD mouse model Neuro HD Incorporates PN backbone chemistry modifications


Slide 18

WVE-003: Clinical trial to leverage experience and learnings in HD Adaptive SAD/MAD design Patients with confirmed manifest HD diagnosis with SNP3 mutation (up to 40 patients planned) Dose escalation and dosing interval guided by independent DSMB Safety and tolerability Biomarkers mHTT NfL wtHTT Clinical trial site activation ongoing Dosing in Phase 1b/2a trial expected to initiate in 2021 Leveraging learnings from PRECISION-HD Starting dose informed by preclinical in vivo models Asuragen assay to improve efficiency of patient identification Drawing from experience of sites from PRECISION-HD1 and PRECISION-HD2 trials SAD: Single ascending doseMAD: Multiple ascending dose mHTT: mutant huntingtinNfL: neurofilament light chain wtHTT: wild-type huntingtin Neuro HD


Slide 19

Three allele-selective HD programs Intend to explore efficacy in early manifest and pre-manifest HD patient populations Neuro HD Potential to address ~80% of HD patient population % Huntington’s Disease Patient Population with SNP SNP1 WVE-120101 SNP2 WVE-120102 SNP3 WVE-003 SNP1 SNP2 SNP1 SNP2 SNP3 ~50% ~50% ~40% ~70%1 ~80%2 +10% of HD patients vs. SNP1 + SNP2 1 Percentage of patient population with SNP1 and/or SNP2 2 Percentage of patient population with SNP1, SNP2 and/or SNP3


Slide 20

WVE-004 Amyotrophic Lateral Sclerosis (ALS) Frontotemporal Dementia (FTD)


Slide 21

C9orf72 repeat expansions: A critical genetic driver of ALS and FTD Normal (non-expanded) Allele < 25 GGGGCC repeats Expanded Allele Sources: DeJesus-Hernandez et al, Neuron, 2011. Renton et al, Neuron, 2011. Zhu et al, Nature Neuroscience, May 2020 Typically 100’s-1000’s of GGGGCC repeats C9orf72 hexanucleotide repeat expansions (GGGGCC) are one of the most common genetic causes of the sporadic and inherited forms of Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD) The C9orf72 repeat expansions also lead to accumulation of repeat-containing transcripts, nuclear sequestration of RNA binding proteins and synthesis of toxic dipeptide-repeat (DPR) proteins  The C9orf72 repeat expansions lead to reduced expression of wild-type C9orf72 and to cellular changes that reduce neuronal viability Neuro C9orf72


Slide 22

C9-ALS and C9-FTD: Manifestations of a clinical spectrum Disease C9 specific US population Mean disease duration Standard of care C9-ALS Fatal neurodegenerative disease  Progressive degeneration of motor neurons in brain and spinal cord ~2,000 3.1 years Significant unmet need despite two approved therapies in US C9-FTD Progressive neuronal atrophy in frontal/temporal cortices Personality and behavioral changes, gradual impairment of language skills ~10,000 6.4 years  No approved disease modifying therapies Two devastating diseases with a shared genetic basis ALS: Amyotrophic lateral sclerosis; FTD: Frontotemporal dementia Sources: Cammack et al, Neurology, October 2019. Moore et al, Lancet Neurology, February 2020 Neuro C9orf72


Slide 23

C9orf72 repeat expansions: Mechanisms of cellular toxicity C9-ALS and C9-FTD may be caused by multiple factors: Insufficient levels of C9orf72 protein Accumulation of repeat-containing RNA transcripts Accumulation of aberrantly translated DPR proteins Recent evidence suggests lowering C9orf72 protein exacerbates DPR-dependent toxicity Sources: Gitler et al, Brain Research, September 2016. Zhu et al, Nature Neuroscience, May 2020 Targeted by Wave ASOs Variant-selective targeting could address multiple potential drivers of toxicity Neuro C9orf72


Slide 24

Normal C9orf72 allele produces three mRNA transcripts (~80% are V2, ~20% are V1 and V3) Pathological allele with expanded repeat leads to healthy V2 and pathological V1 and V3 transcript by-products C9orf72 targeting strategy spares C9orf72 protein WVE-004 targets only V1 and V3 transcripts, sparing V2 transcripts and healthy C9orf72 protein pre-mRNA variants Pathological mRNA products V1 V2 Mis-spliced V1/V3 Stabilized intron1 V3 Disease-causing factors RNA foci Dipeptide repeat proteins (DPRs) GGGGCC expansion Accessible target for variant selectivity WVE-004 reduces Repeat-containing transcripts Neuro C9orf72


Slide 25

PN backbone chemistry modifications: Improved potency among C9orf72-targeting oligonucleotides in vivo Exposure (µg/g) Exposure (µg/g) C9orf72 compounds Spinal cord Cortex PS/PO backbone PS/PO/PN backbone %C9orf72 V3 transcript remaining Mice received 2 x 50 ug ICV doses on days 0 & 7; mRNA from spinal cord and cortex quantified by PCR (Taqman assay) 8 weeks later. Oligonucleotide concentrations quantified by hybridization ELISA. Graphs show robust best fit lines with 95% confidence intervals (shading) for PK-PD analysis. Spinal Cord Neuro C9orf72


Slide 26

WVE-004: Potent and selective knockdown of repeat-containing transcripts in vitro V3 Dose (μM) All V WVE-004 NTC Dose (μM) In vitro activity in C9 patient-derived neurons WVE-004 NTC Dose (μM) IC50:201.7nM In vitro selectivity in C9 patient-derived neurons C9 patient-derived motor neurons were treated with C9orf72 candidate and NTC under gymnotic conditions up to 10uM. Taqman qPCR assays were used to evaluating V3 and all V transcripts. NTC- non-targeting control. Relative fold change C9orf72 V3/HPRT1 1.5 1.0 0.5 0.0 0.001 0.01 0.1 1 10 Relative fold change C9orf72 V3/HPRT1 0.016 0.08 0.4 2 10 0.016 0.08 0.4 2 10 0.016 0.08 0.4 2 10 0.016 0.08 0.4 2 10 1.5 1.0 0.5 0.0 1.5 1.0 0.5 0.0 Relative fold change C9orf72/HPRT1 Neuro C9orf72


Slide 27

WVE-004 demonstrates durable reduction of DPRs in vivo after 6 months in spinal cord and cortex Spinal cord Cortex Full results presented at the 31st International Symposium on ALS/ MND (December 2020) Top: 2 x 50 ug (day 0, day 7) dosed ICV; DPRs measured by Poly-GP MSD assay. *: p≤ 0.05 **: P ≤ 0.01, ***: P ≤ 0.001. ICV: intracerebroventricular; DPR: Dipeptide repeat protein; Bottom: C9 BAC transgenic mice administered PBS or 50 ug WVE-004, ICV, (day 0, day 7). ns: not significant; PBS: phosphate-buffered saline * *** ** 4 12 18 12 18 24 4 24 WVE-004 PBS week 1.5 0.5 0.0 1.0 Relative Poly-GP levels (normalized to PBS) p≤0.0001 4 12 18 12 18 24 4 24 WVE-004 PBS week 1.5 0.5 0.0 1.0 Relative Poly-GP levels (normalized to PBS) ns Relative fold change C9orf72/HPRT1 1.5 0.5 0.0 1.0 WVE-004 PBS ns Relative fold change C9orf72/HPRT1 1.5 0.5 0.0 1.0 WVE-004 PBS Healthy C9orf72 protein relatively unchanged ~6 months after WVE-004 administration Neuro C9orf72


Slide 28

WVE-004: Adaptive SAD/MAD design to optimize dose level and frequency Patients with documented C9orf72 expansion and confirmed ALS, FTD, or mixed phenotype (up to 50 patients planned) Starting dose informed by preclinical in vivo models Dose escalation and dosing interval guided by independent DSMB Key biomarkers of target engagement and neurodegeneration will be assessed PolyGP NfL Key exploratory clinical outcome measures ALSFRS-R and CDR-FTLD Clinical trial site activation ongoing Dosing in Phase 1b/2a trial expected to initiate in 2021 CTA: clinical trial application; NfL: neurofilament light chain; ALSFRS-R: Amyotrophic Lateral Sclerosis Functional Rating Scale; CDRFTLD: Clinical Dementia Scale – frontotemporal lobar degeneration;PolyG: poly glycine-proline; SAD: Single ascending dose; MAD: Multiple ascending dose Neuro C9orf72


Slide 29

WVE-N531 Duchenne muscular dystrophy


Slide 30

WVE-N531 in vitro dose-dependent dystrophin restoration WVE-N531 contains novel PN backbone chemistry modifications Free uptake for 6 days in differentiation media with no transfection agent and no peptide conjugated to the oligonucleotide Demonstrated a dose-dependent increase in dystrophin restoration in DMD patient-derived myoblasts Experimental conditions: Δ45-52 (D45-52) patient myoblasts were treated with oligonucleotide for 6d under free-uptake conditions in differentiation media. Protein harvested in RIPA buffer and dystrophin restoration analyzed by Western Blot. Signal normalized to vinculin loading control and to primary healthy human myotube lysate (pooled from four donors) forming a standard curve in Δ45-52 cell lysate. Western Blot normalized to primary healthy human myoblast lysate Dystrophin protein restoration of up to 71% Neuro DMD


Slide 31

PN chemistry led to overall survival benefit in dKO model  dKO; double knockout mice lack dystrophin and utrophin protein. mdx mice lack dystrophin. Left: Mice with severe disease were euthanized. dKO: PS/PO/PN 150 mg/kg n= 8 (p=0.0018); PS/PO/PN 75 mg/kg n=9 (p=0.00005); PS/PO n=9 (p=0.0024), PBS n=12 Stats: Chi square analysis with pairwise comparisons to PBS using log-rank test PN-containing molecules led to 100% dKO survival at time of study termination PS/PO/PN, 75 mg/kg bi-weekly PBS PS/PO, 150 mg/kg weekly PS/PO/PN, 150 mg/kg weekly 100 75 50 25 0 Survival probability (%) 0 4 8 12 16 20 24 28 32 36 40 Time (weeks) Note: Untreated, age-matched mdx mice had 100% survival at study termination [not shown] Neuro DMD


Slide 32

Clinical trial of WVE-N531 to initiate in 2021 Unmet need in DMD remains high Planned clinical trial designed to evaluate change in dystrophin production, drug concentration in muscle, and initial safety Open-label study; targeting every-other-week administration in up to 15 boys with DMD Potential to apply PN chemistry to other exons if successful CTA submission expected in 1Q 2021 Neuro DMD


Slide 33

Wave’s discovery and drug development platform


Slide 34

Through iterative analysis of in vitro and in vivo outcomes and machine learning-driven predictive modeling, Wave continues to define design principles that are deployed across programs to rapidly develop and manufacture clinical candidates that meet pre-defined product profiles Multiple modalities Silencing | Splicing | ADAR editing DESIGN Unique ability to construct stereopure oligonucleotides with one defined and consistent profile Enables Wave to target genetically defined diseases with stereopure oligonucleotides across multiple therapeutic modalities OPTIMIZE A deep understanding of how the interplay among oligonucleotide sequence, chemistry, and backbone stereochemistry impacts key pharmacological properties SEQUENCE STEREOCHEMISTRY CHEMISTRY Sequence Stereochemistry Chemistry


Slide 35

Sequence Stereochemistry Chemistry PRISM platform enables rational drug design Chemistry R: 2’ modifications OMe, MOE, F, other modifications 5’ 2’ 3’ 5’ 3’ 2’ R X B B X: backbone chemistry Phosphodiester (PO), phosphorothioate (PS), Phosphoramidate diester (PN) Sequence B: bases A, T, C, mC, G, U, other modified bases Stereochemistry Chiral control of any stereocenter 5’ modifications, backbone modifications


Slide 36

Backbone modification (X) Phosphodiester Phosphorothioate Phosphoramidate diester Stereochemistry Not chiral Chiral Chiral Charge Negative Negative Neutral Depiction PRISM backbone modifications Expanding repertoire of backbone modifications with novel PN backbone chemistry Molecule structure illustrative of backbone modification patterns Backbone linkages PS PO PN PO/PS PO/PS/PN Phosphoryl guanidine x-ray structure Stereorandom PS backbone Rp PS backbone Sp PN backbone Sp PN backbone Rp PN backbone Stereorandom


Slide 37

Silencing PN chemistry increases potency in silencing, splicing, and editing preclinical studies Improved knockdown Splicing Editing Improved skipping Ranked by potency of reference PS/PO compound Ranked by potency of reference PS/PO compound Improved editing PS/PO/PN PS/PO (Stereopure) PS/PO (Stereorandom) Concentration (mM) % Editing PS/PO reference compound PS/PN modified compound % Skipping Target knockdown (% remaining) Presented at Analyst & Investor Research Webcast on August 25, 2020; Right: Data from independent experiments


Slide 38

Lead program in Takeda collaboration reinforces potential of PN chemistry in the CNS Single IT dose of 12 mg (n=3) Therapeutic candidate widely distributed across brain and spinal cord ~90% mRNA knockdown one-month following single dose Substantial and widespread target mRNA reduction following single intrathecal dose in NHPs NHPs: Non-human primates; IT: intrathecal  NHPs were administered 12 mg on day 1 via IT bolus injection; tissue samples were collected from 3 NHPs at 28 days post-dose. Target mRNA knockdown 28 days post-dose


Slide 39

PRISM enables optimal placement of backbone stereochemistry Crystal structure confirms phosphate-binding pocket of RNase H binds 3’-SSR-5’ motif in stereopure oligonucleotide – supports design strategy for Wave oligonucleotides ASO/RNA duplex Yellow spheres represent ‘S’ atoms Phosphate binding pocket RNA cleavage site Target RNA Stereopure Oligonucleotide (C9orf72 compound) RNase H + +


Slide 40

Importance of controlling stereochemistry (Rp) (Sp) Top view Side view Yellow spheres represent ‘S’ atomsPS: Phosphorothioate Number of PS linkages in oligonucleotide backbone No. diastereomers 80T 60T 40T 20T 30B 22M 12M 2M 1M 500K 0 0 10 20 30 40 50 Antisense, exon skipping, ssRNAi ADAR oligonucleotide CRISPR guide Stereochemical diversity Exponential diversity arises from uncontrolled stereochemistry


Slide 41

ADAR editing Platform capability and Alpha-1 antitrypsin deficiency


Slide 42

PRISM platform has unlocked ADAR editing A-to-I editing is one of most common post-transcriptional modifications ADAR is ubiquitously expressed across tissues, including liver and CNS ADAR Target RNA I(G) A Edited RNA Oligonucleotide establishes double-stranded RNA complex Oligonucleotide Modification Delivery A: adenosine; I: inosine; G: guanosine; Nishikura, K. A-to-I editing of coding and non-coding RNAs by ADARs. Nat. Rev. Mol. Cell Biol. 2016; Picardi, E. et al. Profiling RNA editing in human tissues: towards the inosinome Atlas. Scientific reports 5, 14941, doi:10.1038/srep14941 (2015). ADAR editing


Slide 43

PRISM enables practical approach to RNA editing without need for viruses or exogenous protein Intracellular Extracellular space Endogenous ADAR Unedited RNA Wave ADAR-editing Oligonucleotides Exogenous ADAR or other base editors Edited RNA Protein release/ expression Delivery vehicles Alternative Base-Editing Systems Edited RNA Genetic construct or foreign protein ADAR editing


Slide 44

Wave platform Fully chemically modified to increase stability in vivo Chirally-controlled backbone to maximize in vitro activity PN backbone chemistry modification improves editing efficiency No requirement for AAV / nanoparticles GalNAc-conjugated for targeted delivery into liver Avoids permanent off-target DNA edits No immunogenicity from exogenous proteins Reduced off-target effects Advantages of Wave ADAR editing platform Sources: Chen Biochemistry 2019 Chemically modified Simplified delivery Endogenous ADAR ADAR editing


Slide 45

ADAR amenable diseases represent a sizeable opportunity Nearly half of known human SNPs associated with disease are G-to-A mutations A-to-I(G) editing could target tens of thousands of potential disease variants1 ~48% C-to-T C-to-G A-to-T C-to-A A-to-C A-to-G Potentially pathogenic human SNPs by base pair corrections >32,000 potentially pathogenic human SNPs2 SNP: single nucleotide polymorphism A: Adenosine I: Inosine G: Guanosine 1ClinVar database 2Gaudeli NM et al. Nature (2017). ADAR editing


Slide 46

RNA editing opens many new therapeutic applications Fix nonsense and missense mutations that cannot be splice-corrected​ Remove stop mutations​ Prevent protein misfolding and aggregation Alter protein processing (e.g. protease cleavage sites) Protein-protein interactions domains Modulate signaling pathways miRNA target site modification​ Modifying upstream ORFs​ Modification of ubiquitination sites Restore protein function Recessive or dominant genetically defined diseases Modify protein function Ion channel permeability Protein upregulation Haploinsufficient diseases Examples: Examples: Examples: ADAR editing


Slide 47

Significant ADAR editing demonstrated in vitro in NHP and primary human hepatocytes NHP: non-human primate; ACTB: Beta-actin; nd= not determined Total RNA was harvested, reverse transcribed to generate cDNA, and the editing target site was amplified by PCR. In vitro dose-response human hepatocytes In vitro dose-response NHP hepatocytes % Editing % Editing ACTB 1 ACTB 2 ACTB 3 ACTB 1 ACTB 2 ACTB 3 ACTB GalNAc-conjugated oligonucleotides with stereopure PN backbone chemistry modifications ADAR editing


Slide 48

Efficient ADAR editing translated in vivo in non-human primate study NHP: non-human primate; ACTB: Beta-actin; Left: 5mg/kg SC: Day 1,2,3,4,5; Liver Biopsy for mRNA (ACTB Editing) & eASO Exposure: Day 7 Up to 50% editing efficiency observed at Day 7, 2 days post last dose Substantial and durable editing out to at least Day 50, 45 days post last dose In vivo editing in NHP following  subcutaneous administration Oligonucleotide quantification in NHP following subcutaneous administration 2 days post last dose 45 days post last dose 2 days post last dose 45 days post last dose ACTB 1 ACTB 2 ACTB 3 ACTB 1 ACTB 2 ACTB 3 Untreated (pre dose) % Editing µg of oligonucleotide per gram of tissue ADAR editing


Slide 49

Editing site RNA editing within ACTB transcript (human hepatocytes) RNA editing within transcriptome (human hepatocytes) Wave ADAR editing oligonucleotides are highly specific Coverage Genome coordinates ACTB C 0% T 100% C 53.8% T 46.2% ACTB Confidence (LOD score) % Editing Mock Editing oligonucleotide Human hepatocytes were dosed with 1um oligonucleotide, 48 hours later RNA was collected and sent for RNA sequencing. RNAseq conducted using strand-specific libraries to quantify on-target ACTB editing and off-target editing in primary human hepatocytes; plotted circles represent sites with LOD>3 ADAR editing


Slide 50

Advancing Wave’s first ADAR editing program in alpha-1 antitrypsin deficiency (AATD) Sources: Strnad 2020; Blanco 2017 AAT: Alpha-1 antitrypsin Most common cause is a single G-to-A point mutation on the “Z” allele ~200K people in US and EU with homozygous ZZ genotype, most common form of severe AATD Approved therapies modestly increase circulating levels of wild-type AAT in those with lung pathology; no therapies address liver pathology Loss of function in lung Gain of function in liver Lack of functional AAT in serum: Insufficient levels to counteract protease levels, e.g., neutrophil elastase Lung damage due to unchecked proteolytic activity and inflammation Other tissues may be affected (e.g., skin) Misfolding of AAT in hepatocytes: Inability to secrete AAT AAT polymerizes in liver Liver damage/cirrhosis Wave’s approach may simultaneously address lung and liver manifestations by using ADAR editing to correct mutation: Increase circulating levels of wild-type AAT protein Reduce aggregation of Z-AAT in liver Retain wild-type AAT physiological regulation Dual pathologies in AATD ADAR editing


Slide 51

SERPINA1 Z allele mRNA editing increases edited AAT protein concentration in vitro AAT (alpha-1 antitrypson); Mouse primary hepatocytes that express SERPINA1 Z allele mRNA were transfected with 25 nanomolar (nM) of SERPINA1 (SA1-1 and SA1-2) targeting antisense oligonucleotides (ASOs) and a control non-targeting (NT) ASO. Media and RNA was collected at 5 days post transfection. AAT protein in media was quantified by Elisa Assay, RNA editing was quantified by RT/PCR/Sanger sequencing. In primary hepatocyte SERPINA1 Z cell model, editing the Z allele mRNA back to wild-type prevents protein misfolding and increases secretion of edited AAT protein from hepatocytes 3-Fold Increase AAT protein concentration in media SERPINA1 Z allele mRNA editing % Z allele mRNA editing AAT protein ng/ml Wild-type AAT protein confirmed by mass spectrometry Function of secreted, edited AAT protein confirmed by neutrophil elastase inhibition assay Model validation and in vivo data expected 1H 2021 Edited AAT protein analysis ADAR editing


Slide 52

Proprietary humanized mouse model developed to support ADAR platform Model validation and in vivo data expected 1H 2021 SERPINA1-Pi*Z/huADAR Protein ü Human ADAR Expressed in all tissues huADAR mouse Protein Pathology ü huADAR Liver pathology, lower huSERPINA1 serum ü SERPINA1 SERPINA1 mouse Protein ü huSERPINA1-Pi*Z Expressed in liver Expression of huADAR in mouse is comparable to expression in human cells  Expression of huADAR restores editing of endogenous targets in primary mouse cell types to levels seen in human primary cell types huADAR mouse model can be crossed with disease specific mouse models to provide model systems for use across Wave’s ADAR editing programs  ADAR editing


Slide 53

Multiple opportunities for ADAR editing in neurology ACTB editing in iCell Neurons ACTB editing in human iCell Astrocytes Concentration (µM) % Editing % Editing Compound 2 (PS / PN) Compound 1 (PS / PN) Compound 3 (PS / PN) EC50: ~200-250nM Gymnotic uptake; Total RNA was harvested, reverse transcribed to generate cDNA, and the editing target site was amplified by PCR and quantified by Sanger sequencing Concentration (µM) ADAR editing hADAR: human ADAR; UGP2: Glucose Pyrophosphorylase 2; 5 mice in each group were injected with PBS or a single 100uG dose on day 0. Animals were necropsied on day 7. RNA was harvested and editing measured by Sanger sequencing. In vivo CNS editing in proprietary hADAR transgenic mouse (1 week)


Slide 54

Ophthalmology


Slide 55

Stereopure oligonucleotides for inherited retinal diseases (IRDs) Wave ophthalmology opportunity Oligonucleotides can be administered by intravitreal (IVT) injection; targeting twice per year dosing Stereopure oligonucleotides open novel strategies in both dominant and recessive IRDs; potential for potent and durable effect with low immune response Successful targeting of MALAT1 is a surrogate for an ASO mechanism of action Widely expressed in many different cell types Only expressed in the nucleus Intravitreal injection Sources: Daiger S, et al. Clin Genet. 2013;84:132-141. Wong CH, et al. Biostatistics. 2018; DOI: 10.1093/biostatistics/kxx069. Athanasiou D, et al. Prog Retin Eye Res. 2018;62:1–23. Daiger S, et al. Cold Spring Harb Perspect Med. 2015;5:a017129. Verbakel S, et al. Prog Retin Eye Res. 2018:66:157-186.; Short, B.G.; Toxicology Pathology, Jan 2008. Ophthalmology


Slide 56

Durable Malat1 knockdown through 9 months with PN backbone chemistry modifications Compound or PBS (1 x 50 ug IVT) was delivered to C57BL6 mice. Relative percentage of Malat1 RNA in the posterior of the eye (retina, choroid, sclera) to PBS-treated mice is shown at 12, 20 and 36 weeks post-single injection. PBS = phosphate buffered saline; NTC= chemistry matched non-targeting control ~50% Malat1 knockdown at 36 weeks in the posterior of the eye PBS NTC PS/PO PS/PN % Malat1 expression Time (weeks) p≤0.01 Ophthalmology


Slide 57

Usher Syndrome Type 2A: a progressive vision loss disorder Autosomal recessive disease characterized by hearing loss at birth and progressive vision loss beginning in adolescence or adulthood Caused by mutations in USH2A gene (72 exons) that disrupt production of usherin protein in retina, leading to degeneration of the photoreceptors No approved disease-modifying therapies ~5,000 addressable patients in US Sources: Boughman et al., 1983. J Chron Dis. 36:595-603; Seyedahmadi et al., 2004. Exp Eye Res. 79:167-173; Liu et al., 2007. Proc Natl Acad Sci USA 104:4413-4418. Oligonucleotides that promote USH2A exon 13 skipping may restore production of functional usherin protein Ophthalmology


Slide 58

Potent USH2A exon 13 skipping with stereopure compound in vitro and ex vivo Oligonucleotides were added to Y79 cells under free-uptake conditions. Exon skipping was evaluated by Taqman assays. USH2A transcripts were normalized to SRSF9. Data are mean±s.d., n=2. Stereorandom: Compound identified in van Diepen et al. 2018. Antisense oligonucleotides for the treatment of eye disease. W02018055134A1. Stereopure: is a stereopure antisense oligonucleotide. Right: Whole NHP were enucleated (n=4) and compounds (1–20 mM) were added to extracted retinas under free-uptake conditions. Exon skipping was evaluated by Taqman assays on RNA. USH2A transcript levels were normalized to SRSF9. Data presented are mean± s.e.m. stereorandom compound is from van Diepen et al. 2018. Antisense oligonucleotides for the treatment of eye disease. W02018055134A1. Compound-1 is a stereopure antisense oligonucleotide. Enhanced potency over a stereorandom reference compound (in vitro) Ophthalmology Target engagement in NHP retinas


Slide 59

Stereopure oligonucleotide elicits dose-dependent exon skipping in NHP eye in vivo Oligonucleotide is complementary to NHP USH2A exon 12* Evaluated 1-week post-single IVT injection Dose-dependent activity of stereopure oligonucleotides Substantial exposure in retina Exon-skipping integrity confirmed by RNA-seq at both doses NTC 150 mg 75 mg 150 mg PBS Stereopure Stereopure USH2A skipping oligonucleotide, PBS or NTC antisense oligonucleotide was delivered to NHP by single IVT injection. One-week post-injection, retina was isolated and exon skipping was evaluated by Taqman assays. USH2A skipped transcript levels were normalized to SRSF9. Data are mean± s.e.m. Stereopure is an USH2A exon-13 skipping stereopure antisense oligonucleotide. PBS, phosphate buffered saline; NTC, non-targeting control; IVT, intravitreal Dose-dependent and specific exon skipping in NHP eye *NHP exon 12 = human exon 13 Ophthalmology


Slide 60

Allele-selective reduction of SNP-containing allele for adRP associated with Rhodopsin P23H mutation Left: Reporter assays on a sequence described in WO2016138353A1. Oligonucleotide and luciferase reporter plasmids (wild-type and mutant RHO) are transfected into Cos7 cells. Cells are harvested after 48 hrs, and relative luminescence is measured. Right: Single IVT injection (1 mL) in mouse Rho P23H mouse model or (150 mL) in human P23H pig model. Eyes collected 1-week post injection for mouse or 2-weeks post injection for pig; RNA isolated; Rho, Hprt1, and Gapdh levels determined by qPCR. Ophthalmology Retinitis pigmentosa (RP): group of rare, genetic eye disorders resulting in progressive photoreceptor cell death and gradual functional loss; currently no cure ~10% of US autosomal dominant RP cases are caused by the P23H mutation in the rhodopsin gene (RHO) Mutant P23H rhodopsin protein is thought to misfold and co-aggregate with wild-type rhodopsin, resulting in a gain-of-function or dominant negative effect in rod photoreceptor cells


Slide 61

THERAPEUTIC AREA / TARGET Milestone Huntington’s disease mHTT SNP2 End of 1Q 2021: PRECISION-HD2 data, including complete 32 milligram cohort, and initial data from OLE trial Huntington’s disease mHTT SNP1 End of 1Q 2021: PRECISION-HD1 data, including complete 16 milligram cohort, and initial data from OLE trial Huntington’s disease mHTT SNP3 2021: Dosing of first patient in clinical trial of WVE-003 ALS and FTD C9orf72 2021: Dosing of first patient in clinical trial of WVE-004 Duchenne muscular dystrophy Exon 53 End of 1Q 2021: CTA submission ADAR editing Multiple 1H 2021: Humanized mouse model validation AATD (ADAR editing) SERPINA1 1H 2021: in vivo AATD data NEUROLOGY HEPATIC Expected upcoming milestones ALS: Amyotrophic lateral sclerosis; FTD: Frontotemporal dementia; AATD: Alpha-1 antitrypsin deficiency Stereopure PN chemistry First clinical compounds with PN chemistry to begin dosing in 2021


Slide 62

Realizing a brighter future for people affected by genetic diseases For more information: Kate Rausch, Investor Relations krausch@wavelifesci.com 617.949.4827