8-K
7 Straits View #12-00 false 0001631574 0001631574 2019-10-07 2019-10-07

 

 

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): October 7, 2019

 

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 7.01 Regulation FD Disclosure.

On October 7, 2019, Wave Life Sciences Ltd. (the “Company”) hosted a Research Day in Boston, Massachusetts and by live webcast, and shared a slide presentation that 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 99.1 to this Current Report on Form 8-K.

The information in this Item 7.01 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 it be deemed incorporated by reference in any filing under the Securities Act of 1933, as amended, or the Exchange Act, except as expressly set forth by specific reference in such a filing.

Item 9.01 Financial Statements and Exhibits.

(d) Exhibits

Exhibit
No.

   

Description

         
 

99.1

   

Research Day Presentation for Wave Life Sciences Ltd. dated October 7, 2019

         
 

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: October 7, 2019

EX-99.1

Slide 1

Analyst and Investor Research Day OCTOBER 7, 2019 BOSTON, MASSACHUSETTS Exhibit 99.1


Slide 2

Welcome Paul Bolno, MD, MBA President and CEO Wave Life Sciences


Slide 3

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 4

Our purpose at Wave Genetic medicines company committed to delivering life-changing treatments for people battling devastating diseases Building a genetic toolbox I II III


Slide 5

Our purpose at Wave Genetic medicines company committed to delivering life-changing treatments for people battling devastating diseases I One tool appears suitable from a distance Building a genetic toolbox for a lifetime of treatment


Slide 6

Our purpose at Wave Genetic medicines company committed to delivering life-changing treatments for people battling devastating diseases Target variability requires a comprehensive toolkit I Building a genetic toolbox for a lifetime of treatment


Slide 7

Our purpose at Wave Genetic medicines company committed to delivering life-changing treatments for people battling devastating diseases Building a genetic toolbox I II III Splicing, silencing, editing Non-viral delivery to nucleus Optimized, stereopure oligonucleotides


Slide 8

Our purpose at Wave Genetic medicines company committed to delivering life-changing treatments for people battling devastating diseases Building a genetic toolbox Designing precision medicines for complex diseases I II III Restoring functional protein Selectively reducing toxic protein Pursuing broad distribution Splicing, silencing, editing Non-viral delivery to nucleus Optimized, stereopure oligonucleotides


Slide 9

Our purpose at Wave Genetic medicines company committed to delivering life-changing treatments for people battling devastating diseases Building a genetic toolbox Designing precision medicines for complex diseases Committed to patients in need I II III Restoring functional protein Selectively reducing toxic protein Pursuing broad distribution Advancing innovative drug development approaches Scaling manufacturing expertise and capacity Building commercial infrastructure Developing novel payor strategies Splicing, silencing, editing Non-viral delivery to nucleus Optimized, stereopure oligonucleotides


Slide 10

Today’s Agenda Gregory Verdine, Ph.D. Co-founder / Board Member | Wave Life Sciences Chandra Vargeese, Ph.D. SVP, Head of Drug Discovery | Wave Life Sciences Elena Cattaneo, Ph.D. University of Milano Frédéric Saudou, M.Sc., Ph.D. Grenoble Institute of Neurosciences (GIN) Chandra Vargeese, Ph.D. SVP, Head of Drug Discovery | Wave Life Sciences Michael Byrne, Ph.D. Director In Vivo Biology & Ophthalmology | Wave Paul Bolno, MD, MBA President & CEO | Wave Life Sciences Chirality Matters in Biology PRISM Biology of Huntingtin (HTT) Biology of Huntingtin (HTT) Advancing HD Portfolio with mHTT SNP3 Lead Inherited Retinal Disease Program: USH2A Closing Remarks PRISM Huntington’s disease Ophthalmology Paul Bolno, MD, MBA President & CEO | Wave Life Sciences Opening Remarks


Slide 11

Chirality Matters in Biology Gregory Verdine, Ph.D. Co-founder and Board Member Wave Life Sciences


Slide 12

Chirality in biology carvone R-carvone S-carvone H Chirality Matters


Slide 13

Stereochemistry Matters in Drugs – Case of Thalidomide Vargesson et al., 2015. Thalomide-induced teratogenesis: History and mechanisms. Birth Defects Res C Embryo Today 105:140-156. ACS Molecule of the week archive: Sept. 1, 2014. Thalidomide was prescribed for the treatment of morning sickness in pregnant women. Between 1957 and 1962, thalidomide caused severe birth defects in >10,000 children. Thalidomide is a mixture of two stereoisomers. One stereoisomer (R) is responsible for the therapeutically beneficial effects. The other (S) isomer causes birth defects. Drugs should be stereochemically pure. (R) (S) Teratogen Anti-emetic Chirality Matters


Slide 14

Oligonucleotides Phosphorothioate (PS) modifications introduce chiral centers Nucleoside Enormous number of permutations exist (2n) à Resulting in thousands of different molecules in every dose Stereopure ASO 1 diastereoisomer 215 = 32,768 diastereoisomers Stereorandom ASO Phosphodiester Stereorandom Rp Sp Phosphorothioate (PS) Chirality Matters


Slide 15

Stereochemical diversity No. PS linkages Antisense, exon skipping, ssRNAi CRISPR guide ADAR oligonucleotide No. diastereomers Exponential diversity arises from uncontrolled stereochemistry (Rp) (Sp) Top view Side view


Slide 16

Mipomersen Iwamoto et al., 2017. Control of phosphorothioate stereochemistry substantially increases the efficacy of antisense oligonucleotides. Nat. Biotechnol. 35:845-851. 524,288 diastereomers Stereopure diastereomers of mipomersen (WV-1-WV-6) Mipomersen Chirality Matters


Slide 17

Overall ASO/RNA/RNase H complex structure Yellow spheres represent ‘S’ atoms + Target RNA + ASO drug ASO/RNA duplex RNase H Phosphate Binding Pocket RNA Cleavage site Chirality Matters


Slide 18

Phosphate binding pocket 1.3 Å resolution RNase H1 Phosphate Binding Pocket 5’ 3’ Rp Sp Sp Scissile Phosphate R179 W225 I239 N240 ASO RNA T181 S223 Chirality Matters


Slide 19

Precision RNase H-mediated RNA degradation In RNase H1 assay, ASOs were pre-annealed to surrogate MALAT1 mRNA (1:1, Cf=5 mM). RNase H (250:1, E:S) was added and quenched with EDTA at the indicated times. Products were quantified, and V0 was calculated from the best-fit line (n=3 per time point). In iCell neurons, 10, 30, 100, 300, 1,000 or 3,000 nM ASO was added to iCell neurons under free-uptake conditions. 4-days post-treatment, RNA was harvested and processed. MALAT1 mRNA expression was determined by qPCR (n=2 per concentration). Control is non-targeting oligonucleotide. In two separate experiments, mice received a single IVT injection of 1 µL in both eyes. One-week post-injection, eyes were enucleated, flash frozen, bisected into anterior and posterior and processed for RNA. MALAT1 mRNA expression was determined by qPCR (each dot represents one eye). Improved rate and amount of cleavage Increased potency in vitro Translatable potency shift in vivo (eye) V0=18 nM/sec V0=8.9 nM/sec Stereorandom Stereopure Control IC50 Stereorandom 2,864 nM Stereopure 120 nM 24- fold 2- fold Stereorandom Stereopure % mRNA Remaining (Malat1/Sfrs9) ~50- fold (µg) Chirality Matters Control


Slide 20

PRISM Chandra Vargeese, Ph.D. SVP, Head of Drug Discovery Wave Life Sciences


Slide 21

PRISM: Wave’s proprietary discovery and drug development platform PRISM Platform progress Applied learnings New modality: ADAR-mediated RNA editing


Slide 22

PRISM Continuous Learning PRISM platform enables rationale drug design


Slide 23

PRISM platform advancing Stereopure Stereorandom ~5-fold improvement in primary screen hit rates Advancement of PRISM platform PRISM All screens used iPSC-derived neurons; Data pipeline for improved standardization


Slide 24

PRISM: Wave’s proprietary discovery and drug development platform PRISM Platform progress Applied learnings New modality: ADAR-mediated RNA editing


Slide 25

Broad tissue distribution and durable target engagement Single IV injection of Wave compounds targeting MALAT1 (human equivalent of 1.6 mg/kg) PBS Compound-1 Compound-3 Gastrocnemius Quadriceps Diaphragm Sciatic nerve Lung Heart PRISM Mice were dosed with a single IV injection (25 mg/kg) of MALAT1-targeting compound, and tissues were assessed for RNA expression 1-, 2-. 4-, and 8-weeks post-dose. Relative percentage of MALAT1 RNA to PBS-treated mice (n=5 per group). MALAT1 RNA levels are normalized to Hprt1.


Slide 26

CNS: Potent and durable targeting with PRISM designed oligonucleotides MALAT1 knockdown in mouse CNS 10-weeks post-dose Compound-2 Broad distribution >80% knockdown of MALAT1 in multiple regions and cell types Knockdown observed 10-weeks after single 100µg dose PRISM Mice received a single 100 mg ICV injection (n=3 per group). Relative fold-change in MALAT1 expression is shown for the indicated tissues 10-weeks post-dose. MALAT1 expression levels are normalized to Hprt1. PBS, phosphate buffered saline; Ctx, cortex; Str, striatum; Cb, cerebellum; Hp, hippocampus; SC, spinal cord. In vivo durability


Slide 27

CNS: Allele-selective silencing of expanded C9orf72 repeat containing transcripts C9orf72 genetic mutations are the strongest genetic risk factor found to date for the more common, non-inherited (sporadic) forms of Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD); GGGGCC repeat drives the formation and accumulation of dipeptide repeat proteins that accumulate in brain tissue Wave’s approach: Selectively silence the GGGGCC repeat containing transcript while minimizing the impact on normal C9orf72 protein Repeat containing transcripts in spinal cord **** ns PBS Lead Candidate PBS Lead Candidate PRISM Experimental description: 2 x 50 ug on day 1 and day 8; mRNA Samples were analyzed using quantitative PCR (Taqman assay), Protein samples were measured by Western Blot. Selective silencing of C9orf72 in vivo (transgenic mouse) Clinical development expected to initiate in 2H 2020 C9orf72 protein in spinal cord Reduction of repeat containing transcripts Protein preservation p<0.0001


Slide 28

PRISM enables resolution of different stereoisomer toxicity profiles Single Rp to Sp shift increases biomarkers for hepatotoxicity Compound-4 and Compound-5 have identical: Sequence Chemical modifications Backbone modifications In vitro potency In vivo potency GalNAc GalNAc Wave Compound-4 Wave Compound-5 Wave Compound-4 Wave Compound-5 PRISM CD1 mice dosed four by 5 mg/kg on days 1, 4, 8, 11; necropsy done day 14


Slide 29

Bright field view Nucleus: Hematoxylin; Light Blue Wave oligo: ViewRNA, Fast Red Nucleus: Hoechst33342; Blue Wave oligo: Fast Red/Cy3; Pink Red 63x oil Fluorescence channel view Z Stack view Stereopure 51, n=82 Stereorandom 5, n=132 # of oligo foci/nuclei 0 50 100 150 200 250 P < 0.001 Stereopure Stereorandom Red Oligonucleotide Blue Nucleus PRISM stereopure oligonucleotides designed to enter the nuclei of cells under free-uptake conditions Free uptake of stereorandom and stereopure oligonucleotides Rapid distribution of stereopure oligonucleotide to muscle in vivo PRISM Experimental conditions: Free uptake of ASOs in 18 hour differentiating human DMD myoblasts (Δ48-50). Data derived from in vivo preclinical research. Experimental conditions: A single dose of stereopure oligonucleotide 30 mg/kg IV was administered to mdx 23 mice. Tissues collected 24 hours post dose and ASO was detected in muscles using ViewRNA. Stereopure oligonucleotides more readily enter the nuclei of cells under free-uptake conditions, which approximates natural delivery in the body


Slide 30

PRISM exon-skipping programs restore significant dystrophin in vitro *Analogs dystrophin (400-427 kDa) vinculin (120 kDa) Marker Mock drisapersen* eteplirsen* suvodirsen WV-isomer 2 WV-isomer 3 Skeletal Muscle Tissue lysates Dystrophin protein restoration of up to 71% in vitro Western Blot normalized to primary healthy human myoblast lysate Dystrophin protein restoration of ~52% in vitro 4Q 2019: Interim clinical dystrophin data readout from OLE expected 2H 2020: Topline clinical data expected PRISM Experimental conditions: DMD protein restoration by western blot in patient-derived myotubes with no transfection reagents. Free uptake at 10 µM concentration (left panel) of each compound. Clear dose effect for WVE-N531 (right panel). Suvodirsen (Exon 51) WVE-N531 (Exon 53)


Slide 31

PRISM: Wave’s proprietary discovery and drug development platform PRISM Platform progress Applied learnings New modality: ADAR-mediated RNA editing


Slide 32

RNA-editing can be used for several therapeutic applications and supplement Wave’s existing modalities Silence protein expression Reduce levels of toxic mRNA/protein ü ü Alter mRNA splicing Exon skipping/inclusion/ restore frame ü ü Fix nonsense mutations that cannot be splice-corrected Restore protein expression ü Fix missense mutations that cannot be splice-corrected Restore protein function ü Modify amino acid codons Alter protein function ü Remove upstream ORF Increase protein expression ü Strategy Therapeutic Application Silencing Splicing RNA Editing Treatment Modality PRISM I (G): ADAR converts A>I, I is recognized as G by all cellular machinery; ADAR: Adenosine Deaminase Acting on RNA


Slide 33

Structure of ADAR deaminase domain bound to dsRNA substrate Using PRISM to unlock ADAR-mediated RNA editing Target mRNA Oligonucleotide ADAR makes multiple contacts with oligonucleotide backbone, sugar and bases Using PRISM platform, rationally designed and screened oligonucleotides to optimize: 2' sugar chemistry Backbone chemistry and stereochemistry Size and structure Modified nucleobases ~1,000 RNA editing oligonucleotides tested over the last year to develop SAR for editing format PRISM Structure adapted from Matthews et al., Nat Struct Mol Biol. (2016); SAR = structure-activity relationship; ADAR: Adenosine Deaminase Acting on RNA; dsRNA = double-stranded RNA


Slide 34

Wave’s ADAR approach has several advantages over existing technologies Require AAV or lipid nano particle delivery Require exogenous protein (e.g. CAS13 or chimeric ADAR) Free uptake into tissues Uses endogenous ADAR for editing Use unmodified RNA Fully chemically-modified stereopure oligonucleotides Stability Delivery Editing PRISM Existing RNA editing technologies Wave’s RNA editing platform Single oligonucleotide through free uptake is sufficient for editing


Slide 35

RNA-editing with endogenous ADAR achieved across multiple primary human cell types Stereochemistry significantly increases editing across all cell lines tested, especially for gymnotic delivery GalNAc-conjugated fully-modified stereopure oligonucleotide can be used for targeted editing in hepatocytes; in vitro experiments suggest an EC50 of ~100nM in primary hepatocytes In vivo editing with fully-modified stereopure oligonucleotide studies underway Hepatocytes (GalNAc-Mediated Uptake) Hepatocytes (Gymnotic Uptake) Bronchial Epithelial Cells (Gymnotic Uptake) Stereopure oligonucleotide Stereorandom oligonucleotide No editing detected Stereopure oligonucleotide Stereorandom oligonucleotide PRISM In vivo editing data expected in 2020 Editing UAG Site in Actin mRNA in Primary Human Cell Lines


Slide 36

Portfolio Paul Bolno, MD, MBA President and CEO Wave Life Sciences


Slide 37

THERAPEUTIC AREA/MODALITY TARGET DISCOVERY CANDIDATE CLINICAL REGISTRATION ESTIMATED U.S. PREVALENCE* PARTNER Duchenne muscular dystrophy Exon-skipping Suvodirsen Exon 51 ~2,000 WVE-N531 Exon 53 ~1,250 Exons 44, 45, 52, 54, 55 ~3,000 Neuromuscular diseases Multiple Huntington’s disease Allele – selective silencing WVE-120101 mHTT SNP1 ~10,000 / ~35,000 Takeda 50:50 option WVE-120102 mHTT SNP2 ~10,000 / ~35,000 Takeda 50:50 option mHTT SNP3 ~8,000 / ~30,000 Takeda 50:50 option ALS and FTD Allele – selective silencing WVE-C092 C9orf72 ~1,800 (ALS) ~7,000 (FTD) Takeda 50:50 option Spinocerebellar ataxia 3 Silencing ATXN3 ~4,500 Takeda 50:50 option CNS diseases Multiple† Takeda milestones & royalties Retinal diseases USH2A and multiple Metabolic liver diseases Silencing Multiple Pfizer milestones & royalties MUSCLE CNS OPHTHALMOLOGY HEPATIC U.S. A.A. filing planned in 2H 2020 pending dystrophin data OLE and Phase 2/3 Phase 1b/2a Phase 1b/2a Pipeline spanning multiple modalities, novel targets PRISM *Estimates of U.S. prevalence and addressable population by target based on publicly available data and are approximate; for Huntington’s disease, numbers approximate manifest and pre-manifest populations, respectively. †During a four-year term, Wave and Takeda may collaborate on up to six preclinical targets at any one time. A.A.: Accelerated approval; ALS: Amyotrophic lateral sclerosis; FTD: Frontotemporal dementia; CNS: Central nervous system


Slide 38

Biology of Huntingtin (HTT) Elena Cattaneo, Ph.D. University of Milano Frédéric Saudou, M.Sc., Ph.D. Grenoble Institute of Neurosciences (GIN)


Slide 39

Prof. of pharmacology, director of Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases Director of UniStem (Centre for Stem Cell Research of the University of Milan) Earned PhD in biotechnology applied to pharmacology at University of Milan Completed first post-doc at MIT under supervision of Prof. Ronald McKay – studied neural stem cell differentiation associated with neurodegenerative conditions Learned strategies for stem cell grafting at Lund University in the lab of Prof. Anders Björklund Returned to University of Milan in 1995 as a researcher Appointed associate professor in 2001, full professor in 2003 Today her lab focuses on molecular pathophysiology of HD and mechanisms regulating striatal neurodegeneration They are identifying cells, molecules, pathways that are suitable for therapeutic application to slow or prevent the disease In 2013, was appointed Senator for life by President Giorgio Napolitano on account of her scientific and social merit Prof. at University Grenoble Alpes & CHU, director of Grenoble Institute of Neuroscience (GIN) Group leader of the team ‘Intracellular Dynamics and Neurodegeneration’ Director of the Grenoble Center of Excellence in Neurodegeneration (COEN-GREEN) Undertook his thesis at the University of Strasbourg with Prof. René Hen on serotonin receptors Completed first post-doc in Strasbourg with Prof. Jean-Louise Mandel in human genetics Completed second post-doc at Harvard Medical School with Prof. Michael Greenberg on neuronal signaling In 2000, moved back to France to lead research team at the Institut Curie; became director of department in 2010 Research team moved to Grenoble in December 2014; major focus is understanding huntingtin function, dysfunction in intracellular trafficking to investigate pathogenic mechanisms In 2014, received the Richard Lounsbery prize for medicine and biology from the French and US national academies of Science Elena Cattaneo, Ph.D. University of Milano Frédéric Saudou, M.Sc., Ph.D. Grenoble Institute of Neurosciences (GIN) Huntington’s disease


Slide 40

[Placeholder] Elena Cattaneo slides Huntington’s disease


Slide 41

Huntington’s disease [Placeholder] Frédéric Saudou slides


Slide 42

Advancing HD Portfolio with mHTT SNP3 Chandra Vargeese, Ph.D. SVP, Head of Drug Discovery Wave Life Sciences


Slide 43

wt SNP1 SNP3 SNP2 Huntington’s disease Allele-selective silencing Aims to lower mHTT transcript while leaving healthy HTT relatively intact RNase H and ASO:RNA Allele-selectivity possible by targeting SNPs associated with expanded long CAG repeat in HTT gene


Slide 44

Broadening reach in Huntington’s disease with SNP3 development program Due to overlap, ~80% of the total HD patient population carry SNP1 and/or SNP2 and/or SNP3 In vivo models for SNP3 available for preclinical development SNP3 % Huntington’s Disease Patient Population with SNP SNP1 SNP2 SNP3 SNP1 SNP2 SNP1 SNP2 SNP3 ~50% ~50% ~40% ~70% ~80% +10% of HD patients vs. SNP1 + SNP2 Huntington’s disease Predicted patient coverage calculated from published phasing data from Canadian HD patients (Carroll et al., 2011)​​


Slide 45

SNP3 program Potency in homozygous iCell neurons as compared to pan-silencing compound Allele-selectivity in vitro as compared to pan-silencing compound Biochemical assay Heterozygous patient neurons Target engagement and durability in vivo in BACHD models Huntington’s disease


Slide 46

Potent mutant HTT knockdown activity Greater knockdown of mutant HTT as compared to pan-silencing compound Wave allele-selective compounds are more potent than pan-silencing RG6042 analog in patient-derived neurons ~7-fold shift Pan-silencing RG6042 analog Wave SNP3 Compound-1 Wave SNP3 Compound-2 Huntington’s disease HTT mRNA remaining in iCell neurons (homozygous for SNP) incubated with the indicated ASO under free-uptake conditions. Data show mean ± sem (n=4). Homozygous iCell Neurons


Slide 47

Stereopure oligonucleotides are selective in vitro Stereopure isomers targeting a SNP variant promote RNase H-mediated degradation of mutant HTT while sparing wild-type HTT Biochemical RNase H assays RNase H experiments performed with synthetic RNA substrates corresponding to mHTT and wtHTT variants (S:E = 100:1; n=2). Percentage of the indicated full-length RNA substrate remaining over time is plotted for the stereopure SNP3 Compound-1 (left) and stereopure SNP3 Compound-2 (right). Abbreviations: S, substrate; E, enzyme. Wave SNP3 Compound-1/mHTT Wave SNP3 Compound-1/wtHTT Time (min) % RNA substrate remaining Wave SNP3 Compound-2/mHTT Wave SNP3 Compound-2/wtHTT % RNA substrate remaining Time (min) Huntington’s disease


Slide 48

Demonstration of allele-selective silencing Stereopure compounds selectively deplete mutant HTT mRNA Neurons were derived from GM21756 patient-derived fibroblasts (heterozygous for SNP) and treated with 2.2 mM (left) or 20 mM (right) of the indicated ASO under gymnotic conditions for 7 days. RNA was quantified and normalized to TUBB3. Data are mean ± sem (n=3). Percentage of remaining wtHTT and mHTT mRNA is indicated. No loss of selectivity with increasing concentrations [20 µM] PBS PBS Pan-silencing RG6042 analog Pan-silencing RG6042 analog Wave SNP3 Compound-1 Wave SNP3 Compound-2 Wave SNP3 Compound-1 Wave SNP3 Compound-2 [2 µM] Huntington’s disease


Slide 49

In vivo model to assess target engagement and durability BACHD mouse model Expressed transcript includes SNP3 variant that Wave compounds are targeting Model is homozygous for mutant HTT with SNP3 (only has one type of HTT) Over-expresses mHTT (multiple gene copies) No ability to assess allele selectivity Oligonucleotide concentration in tissues Achieved good tissue exposure over 12-weeks in BACHD cortex and striatum Oligonucleotide or PBS (3 x 100 µg ICV) was delivered to BACHD mice. Oligonucleotides were quantified by ELISA. Tissue exposure over time CORTEX STRIATUM Pan-silencing RG6042 analog Wave SNP3 Compound-1 Wave SNP3 Compound-2 Huntington’s disease


Slide 50

Durable in vivo mutant HTT knockdown with stereopure SNP3 compounds Knockdown persists for 12 weeks Oligonucleotide or PBS (3 x 100 mg ICV) was delivered to BACHD mice. Relative percentage of HTT/TUBB3 mRNA in cortex with respect to levels in PBS-treated mice is shown at 2-12 weeks post-injection. Statistics: All oligo treatment groups are statistically significantly different from PBS; One-way ANOVA ****, P≤0.0001. Wave SNP3 Compound-1 and Compound-2 are also significantly different from RG6042 analog at 8 and 12 weeks ***, P<0.005; **P=0.001. Relative percentage mHTT expression Relative percentage mHTT expression BACHD model only has mutant HTT (no wildtype HTT) PBS Pan-silencing RG6042 analog Wave SNP3 Compound-1 Wave SNP3 Compound-2 PBS Pan-silencing RG6042 analog Wave SNP3 Compound-1 Wave SNP3 Compound-2 BACHD Cortex BACHD Striatum ** **** *** **** **** **** Huntington’s disease


Slide 51

Lead Inherited Retinal Disease Program: USH2A Michael Byrne, Ph.D. Director In Vivo Biology and Ophthalmology Wave Life Sciences


Slide 52

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 Lead program USH2A Ophthalmology 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.


Slide 53

Stereopure compounds durably deplete MALAT1 for 9 months in vivo ~50% MALAT1 knockdown at 9 months in the posterior of the eye Ophthalmology Compound or PBS (1 x 50 mg 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. Statistics: Compound-2 Malat1 levels are significantly different from NTC at 36 weeks ***, P<0.001; **** P<0.0001, respectively. PBS = phosphate buffered saline; NTC= chemistry matched non-targeting control; Compound-1 and Compound-2 are stereopure MALAT1-targeting antisense oligonucleotide. Minimal exposure required extended duration PBS NTC Compound-1 Compound-2 Time (weeks) PBS NTC Compound-1 Compound-2 Time (weeks) **** ***


Slide 54

Stereopure compound induces potent and durable MALAT1 knockdown in the eye Ophthalmology Oligonucleotide or PBS (1 x 450 mg IVT) was delivered to NHP. Relative percentage of MALAT1 RNA in the retina to PBS-treated is shown at 1 week, 2 and 4 months, post-single injection. Compound-1 is a stereopure MALAT1-RNA-targeting antisense oligonucleotide. 90% knockdown of MALAT1 in NHP Retina Compound-1 detected in NHP Retina 4-months post-dose Photoreceptors In vivo duration of effect in the NHP Retina >90% knockdown maintained for 4 months PBS Compound-1 Cell Nuclei Compound-1 MALAT1 RNA


Slide 55

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 Ophthalmology 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


Slide 56

Productive USH2A exon 13 skipping with stereopure compound Ophthalmology Compounds 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. Primers mapping to exons 8 and 17 were used to amplify region containing skipped exon. RNA-Seq was performed on the miSEQ platform. Reference Compound: van Diepen et al. 2018. Antisense oligonucleotides for the treatment of eye disease. W02018055134A1. Compound-1 is a stereopure antisense oligonucleotide. Exon skipping in Y79 cells Gel shift & RNA-seq confirm productive exon skipping PBS Compound-1 4-fold shift in potency Exon 13 reads decreased with Compound-1 PBS Compound-1 Exon 8-17 Exon 13 (-642 nucleotides)


Slide 57

Potent USH2A exon skipping ex vivo in NHP and human retinas Ophthalmology Whole NHP and human eyes were enucleated (n=4 and n=2, respectively) and compounds (1–20 µM) 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. Compound-1 is a stereopure antisense oligonucleotide. Target engagement in NHP (left) and human (right) retinas PBS NTC Compound-1 20 20 10 5 1 [µM] PBS NTC Compound-1 20 20 10 [µM]


Slide 58

Autosomal dominant retinitis pigmentosa (adRP) associated with Rhodopsin P23H mutation Retinitis pigmentosa (RP) is a group of rare, genetic disorders of the eye resulting in progressive photoreceptor cell death and gradual functional loss Currently no cure for RP ~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 ~1,800 addressable patients in US Ophthalmology Ferrari et al., Current Genomics. 2011;12:238-249. Allele-selective reduction of the mutant P23H allele while maintaining the wild type rhodopsin allele may prevent further cell loss


Slide 59

adRP associated with Rhodopsin P23H mutation Stereopure oligonucleotides achieve allele-selective reduction of SNP-containing allele Reporter assays on a Wave stereopure sequence as well as a sequence described in WO2016138353A1: ASO and luciferase reporter plasmids (wild-type and mutant rhodopsin) are transfected into Cos7 cells. 48-hours later, cells are harvested, and relative luminescence is measured. Ophthalmology Stereorandom Stereopure Collaborations in place for evaluation in transgenic human Rho P23H pig model In vivo Stereopure compound is allele selective compared with stereorandom


Slide 60

Summary Wave stereopure compounds induce potent and durable MALAT1 knockdown in the eye USH2A is Wave’s lead ophthalmology program Productive USH2A exon 13 skipping in cellular models Confirmed skipping at the sequence level Potent exon skipping demonstrated ex vivo in NHP and human retinas USH2A in vivo studies ongoing Discovery work underway for second ophthalmology program (RHO P23H) Ophthalmology IND-enabling studies for USH2A candidate expected to begin in 2020


Slide 61

Conclusion Paul Bolno, MD, MBA President and CEO Wave Life Sciences


Slide 62

Q&A


Slide 63

Analyst and Investor Research Day OCTOBER 7, 2019 BOSTON, MASSACHUSETTS