Fitusiran Investigational RNAi Therapeutic for the Treatment of Hemophilia and Rare Bleeding Disorders. Monday, August 22, 2016

Similar documents
The Combined Use of Bypassing Agents with Antithrombin Reduction in Plasma of Hemophilia A and B Patients with Inhibitors

Welcome to R&D Day! Christine Lindenboom VP, Investor Relations & Corporate Communications

Supplementary Appendix

Agenda. Welcome. Introduction. Review of Interim ALN-CC5 Phase 1/2 Data. Q&A Session

WFH 2014 World Congress Medical Free Paper 8 ALN AT3: An Investigational RNAi Therapeutic Targeting Antithrombin for the Treatment of Hemophilia

Alnylam Pharmaceuticals R&D Day

2015 Goring Coagulation Conference

AT Reduction Corrects Thrombin Generation in Samples From Hemophilia A & B Patients With Inhibitors

Stifel Nicolaus Healthcare Conference. September 2012

CATALYST BIOSCIENCES. Corporate Overview. 9 April 2019

Disclosure. Hemophilia: The Royal Treatment. Objectives. Background. History of Hemophilia. Epidemiology 1/4/2018

Alnylam Pharmaceuticals

CATALYST BIOSCIENCES. Corporate Overview. 12 February 2019

uniqure Announces First Clinical Data From Second Dose Cohort of AMT-060 in Ongoing Phase I/II trial in Patients with Severe Hemophilia B

5 December 2016 ASH San Diego, CA

Together, all nine participants have reduced infusions of factor IX concentrates by 99 percent over cumulative 1,650 days

Future prospects for the treatment of haemophilia

Prior Authorization Criteria Hemophilia/Blood Factor Products

EVOLUTION OF INHIBITOR TREATMENT

Pre-Clinical Evaluation of ALN-AAT to Ameliorate Liver Disease Associated with Alpha-1 Antitrypsin Deficiency

Updates in Hemophilia: Making Headway in the Multidisciplinary Setting

Abdulkareem Almomen, MD, FRCPC, Professor of Medicine-Hematology, King Saud University & Blood and Cancer Center, Riyadh Jeddah, 24 February 2018

Introduction KEY POLICY IMPLICATIONS SUMMARY A LOOK AT EMICIZUMAB FOR FOR HEMOPHILIA A WITH A INHIBITORS WITH INHIBITORS HEMOPHILIA A

Medical Policy An independent licensee of the Blue Cross Blue Shield Association

Launches The Many Faces of Bleeding Disorders educational video podcast together with the World Federation of Hemophilia

Roche s emicizumab showed positive results in phase III studies (HAVEN 1 and HAVEN 2) in haemophilia A with inhibitors

Alnylam Pharmaceuticals R&D Day. July 11, 2013

Fourth Quarter and Full Year Financial and Operational Results Conference Call March 21, 2018

Evaluation of AMG 531 Efficacy in Splenectomized Patients with Chronic ITP in a Randomized Placebo- Controlled Phase 3 Study

Factor VIII Concentrate Factor IX Complex (Coagulation Factors, II, VII, IX, X) Concentrate

Media Release. CHMP recommends EU approval of Roche s Hemlibra for people with haemophilia A with inhibitors. Basel, 26 January 2018

Genentech Contacts: Media: Tara Cooper (650) Investor: Kathee Littrell (650)

Agios Pharmaceuticals, Inc.

First Quarter 2018 Financial Results. May 8, 2018

Hepatocyte-Targeted RNAi Therapeutics: A Reproducible and Modular Platform for Innovative Medicines

UP AND COMING ANTICOAGULANTS AND HEMOSTASIS AGENTS

Results to be Presented at LDN WORLD Symposium in February Initiation of Repeat-Dose Pompe Study Anticipated in 3Q13

Catalyst Biosciences. Essential Medicines for Hemophilia Greater Convenience Superior Outcomes. 21 June 2018

TIDES 2014 GalNAc-siRNA with Enhanced Stabilization Chemistry: ESC-GalNAc-siRNA. May 14, 2014 Muthiah Manoharan

APOLLO Phase 3 Study of Patisiran Topline Results

DOACs Can Be Reversed!

Sanofi to Acquire Bioverativ A Strategically and Financially Compelling Acquisition in Specialty Care

Media Release. Roche announces positive interim results for emicizumab in phase III study of children with haemophilia A. Basel, 17 April 2017

Measurement of Thrombin Generation in Conditions of Low Antithrombin

iccnet CHSA Clinical Protocol - HEPARIN

Hemophilia and Gene Therapy

trial. Key trial data points:

Total urinary GAGs declined by 51%, dermatan sulfate by 32%, and heparan sulfate by 61% in Cohort 2 at 16 weeks

Corporate Overview. Douglas Fambrough, President and CEO Cowen and Company 39 th Annual Health Care Conference March 12, DICERNA

Second Quarter 2016 Financial Results. August 4, 2016

Treating and Preventing Infectious Disease. November 2011 Nasdaq: INHX

A Patient-Centered Approach to Understanding the Burden of Inhibitors

December 8 December 14, 2017

J.P. Morgan 36 th Annual Healthcare Conference. January 10, 2018

INTERMUNE ANNOUNCES TOP-LINE RESULTS OF PHASE 1B TRIAL OF ITMN-191 (R7227) AND PROVIDES PROGRAM UPDATE

Media Release. Roche s Hemlibra continued to substantially reduce bleeds in people with haemophilia A with inhibitors. Basel, 09 December 2017

Robust and Durable Target Silencing in the CNS with sirna Conjugates

35 th Annual J.P. Morgan Healthcare Conference. Jean-Jacques Bienaimé Chairman and Chief Executive Officer BioMarin Pharmaceutical Inc.

Corporate Overview. January 2019 Douglas Fambrough, CEO

SYNOPSIS. Clinical Study Report for Study CV Individual Study Table Referring to the Dossier

Bevyxxa (betrixaban) NEW PRODUCT SLIDESHOW

The Role of Disease Foundations in Moving Gene Transfer Forward

Spark Therapeutics, Inc.

Corporate Overview. November Alnylam Pharmaceuticals, Inc.

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) DRAFT GUIDELINE ON THE CLINICAL INVESTIGATION OF RECOMBINANT FACTOR VIII AND IX PRODUCTS

Alnylam Pharmaceuticals ALN-AT3 for Treatment of Hemophilia August 7, 2014

Catalyst Biosciences

Hepatocyte-Targeted RNAi Therapeutics: A Reproducible and Modular Platform for Innovative Medicines

New Anticoagulants Linda Liu, M.D.

Haemophilia care in Europe: a survey of 19 countries

Clinical Policy: Humate-P (Antihemophiliac Factor/von Willebrand Factor Complex Human) Reference Number: CP.MP.404

January (San Francisco, CA) January 8, 2018

Mark Crowther, MD, MSc, FRCPC. Co-authors. Professor of Medicine, McMaster University, Canada

70% not diagnosed, 75% not treated Many die in childhood or grow-up severely disabled

Corporate Overview. March 2015

In Hospital Bleeding Management

Media Release. CHMP recommends EU approval of Roche s Hemlibra for people with severe haemophilia A without factor VIII inhibitors

Bayer R&D Investor Day 2005

FDA grants Priority Review to Roche s Hemlibra for people with haemophilia A without factor VIII inhibitors

Patients Perspective in Plasma Products (Focus on Hemophilia)

Patients Perspective in Plasma Products (Focus on Hemophilia)

This presentation contains forward-looking statements within the meaning of the "safe harbor" provisions of the Private Securities Litigation Reform

Deep dive into anticoagulant reversal In 20 minutes so we won t dive too deep. Mark Crowther on behalf many Some slides modified from other sources

1.0 Abstract. Palivizumab P Study Results Final

Corporate Presentation OCTOBER 2018

Second Quarter 2017 Financial Results. August 8, 2017

Platform Advances in RNAi Therapeutics. RNAi Roundtable August 23, 2017

Media Release. Roche to present new phase III data for Hemlibra in people with haemophilia A at the World Federation of Hemophilia 2018 World Congress

Factor Concentrates. More is better? Alexander Duncan MD Emory Medical Labs

This presentation contains forward-looking statements within the meaning of the "safe harbor" provisions of the Private Securities Litigation Reform

Corporate Overview. May 2015

Future prospects for haemophilia therapy

Corporate Presentation JANUARY 2019

Chimerix Announces First Quarter 2017 Financial Results

An Approach to bleeding

Anti- THrombosis with Enoxaparin in intubated Adolescents

Corporate Overview. October 1, 2018 Cantor Douglas Fambrough, CEO

Stealth BioTherapeutics Mission:

Transcription:

Fitusiran Investigational RNAi Therapeutic for the Treatment of Hemophilia and Rare Bleeding Disorders Monday, August 22, 216 1

Agenda Welcome Josh Brodsky, Associate Director, Investor Relations & Corporate Communications Introduction Akin Akinc, Ph.D., Vice President, General Manager, Fitusiran Overview of Hemophilia and Patient Perspective Brian O Mahony, Chief Executive, Irish Haemophilia Society Ltd. and person living with severe hemophilia B Fitusiran Program Akin Akinc, Ph.D., Vice President, General Manager, Fitusiran Benny Sorensen, M.D., Ph.D., Senior Director, Clinical Research Q&A Session 2

Reminders Event will run for approximately 75 minutes Q&A Session at end of presentation Submit questions at bottom of webcast screen Questions may be submitted at any time Replay, slides and audio available at www.alnylam.com 3

Alnylam Forward Looking Statements This presentation contains forward-looking statements, within the meaning of Section 27A of the Securities Act of 1933 and Section 21E of the Securities Exchange Act of 1934. There are a number of important factors that could cause actual results to differ materially from the results anticipated by these forwardlooking statements. These important factors include our ability to discover and develop novel drug candidates and delivery approaches and successfully demonstrate the efficacy and safety of our product candidates; pre-clinical and clinical results for our product candidates; actions or advice of regulatory agencies; delays, interruptions or failures in the manufacture and supply of our product candidates; our ability to obtain, maintain and protect intellectual property, enforce our intellectual property rights and defend our patent portfolio; our ability to obtain and maintain regulatory approval, pricing and reimbursement for products; our progress in establishing a commercial and ex-united States infrastructure; competition from others using similar technology and developing products for similar uses; our ability to manage our growth and operating expenses, obtain additional funding to support our business activities and establish and maintain business alliances; the outcome of litigation; and the risk of government investigations; as well as those risks more fully discussed in our most recent quarterly report on Form 1-Q under the caption Risk Factors. If one or more of these factors materialize, or if any underlying assumptions prove incorrect, our actual results, performance or achievements may vary materially from any future results, performance or achievements expressed or implied by these forward-looking statements. All forward-looking statements speak only as of the date of this presentation and, except as required by law, we undertake no obligation to update such statements. 4

Agenda Welcome Josh Brodsky, Associate Director, Investor Relations & Corporate Communications Introduction Akin Akinc, Ph.D., Vice President, General Manager, Fitusiran Overview of Hemophilia and Patient Perspective Brian O Mahony, Chief Executive, Irish Haemophilia Society Ltd. and person living with severe hemophilia B Fitusiran Program Akin Akinc, Ph.D., Vice President, General Manager, Fitusiran Benny Sorensen, M.D., Ph.D., Senior Director, Clinical Research Q&A Session 5

RNAi Therapeutics New Class of Innovative Medicines Harness natural pathway Catalytic mechanism Silence any gene in genome Upstream of today s medicines Clinically proven approach 6

Alnylam Strategic Therapeutic Areas (STArs) Investigational pipeline focused in 3 STArs Genetic Medicines RNAi therapeutics for rare diseases Cardio-Metabolic Diseases RNAi therapeutics for dyslipidemia, NASH, type 2 diabetes, hypertension, and other major diseases Hepatic Infectious Diseases RNAi therapeutics for major liver infections beginning with hepatitis B & D 7

Alnylam Development Pipeline 8

Alnylam Development Pipeline 9

Agenda Welcome Josh Brodsky, Associate Director, Investor Relations & Corporate Communications Introduction Akin Akinc, Ph.D., Vice President, General Manager, Fitusiran Overview of Hemophilia and Patient Perspective Brian O Mahony, Chief Executive, Irish Haemophilia Society Ltd. and person living with severe hemophilia B Fitusiran Program Akin Akinc, Ph.D., Vice President, General Manager, Fitusiran Benny Sorensen, M.D., Ph.D., Senior Director, Clinical Research Q&A Session 1

Haemophilia and Rare Bleeding Disorders Brian O Mahony Chief Executive, Irish Haemophilia Society Ltd. and person living with severe hemophilia B 11

Haemophilia A FVIII deficiency 15 per million males 1.5 per 1, males 8% of Haemophilia 12

Haemophilia B FIX Deficiency 28 per million males.28 per 1, males 2% of Haemophilia 13

Inheritance of Haemophilia Sex linked Inheritance X chromosome Carrier mother Sons 5% chance, haemophilia Daughters 5% chance, carriers Man with Haemophilia Sons, no haemophilia Daughters, obligatory carriers 3% Spontaneous Mutation 14

Haemophilia - Severity Severe < 1% Moderate 1 to 5 % Mild 5 to 4% Severe - spontaneous bleeds 15

Von Willebrands Type 1 Type 2A Type 2B Type 2N Type 3.1% of Population 16

Von Willebrands Chromosome 12 Affects males, females Females often more symptomatic Severity Proportion Treatment Type 1 Mild 75% DDAVP Type 2 Moderate 2-25% DDAVP, Concentrate Type 3 Severe 1:5, Concentrate 17

Bleeding Aura Pain, Heat, Swelling Damage Early treatment essential 18

Incidence of different sites of bleeding Haemarthrosis: 7-8% Muscle/soft tissue: 1-2% Other major bleeds: 5-1% Central nervous system bleeds: <5% 19

Incidence of bleeding into joints Target Joints Knee 45% Elbow 3% Ankle 15% Shoulder 3% Wrist 3% Hip 2% Other 2% 2

The musculosceletal consequences of undertreatment 21

22

Treatment Principles Prevention of bleeding should be the goal Treat acute bleeds early Home therapy early treatment Severe bleeds hospital Avoid aspirin, Intramuscular injections, non steroidal anti inflammatory agents On demand treatment when bleeding occours Prophylaxis- prevents spontaneous bleeds- maintain Factor level >1% at all times 23

Development of Replacement Therapy 196 s: Plasma, Cryoprecipitate 197 s : Plasma derived clotting Factor concentrates (CFCs) 1985-88 : Virally inactivated plasma derived CFCs 1994: Recombinant CFCs 214: Extended half life CFCs 24

25

26

27

Self-infusion Video 28

Current Treatment Options Cryoprecipitate, Plasma - only in absence of factor concentrates as an option Plasma derived factor concentrates FVIII, FIX,VWD and rare bleeding disorders Recombinant Factor concentrates FVIII, FIX and FXIII DDAVP- mild haemophilia Extended half life factor concentrates- FVIII, FIX 29

Personal Patient Journey 3

EHC Strategic objectives 214-217 1. Support and empower NMOs 2. Promote access to optimal treatment and comprehensive care for people with Haemophilia, VWD and RBDs 3. Ensure constructive engagement with key stakeholders 4. Increase influence on European policymaking environment 5. Ensure good governance and sustainability 31

NMO support NMO & multi-stakeholder workshops Economics and HTAs New Technologies Tenders and Procurement Youth Development European Inhibitor Program Annual Leadership conference EU Policy Reports for NMOs, 4 x year Annual Scientific conference 32

Treatment and care Collect and publish data annually 215: Haemophilia care in Europe (37 countries ) 214: Tenders and procurement systems in Europe ( 37 countries) 216; Hepatitis C and Haemophilia Certification of EHCCCs/EHTCs Medical Advisory Group- statements on important issues Significant input into drafting of Council of Europe recommendations on haemophilia 33

EHC data Collection and Publication Brian O Mahony, 215 34

FVIII use per capita vs GDP 214 data 3 IU/capita Brian O Mahony, Unpublished data 35

Changes in FVIII use between 28 and 214 Brian O Mahony, Unpublished data 36

Stakeholders, Policymaking Environment Round Tables of Stakeholders European Parliament hosted 216: Inhibitors, HCV, Ageing, Outcome Measures Representation EMA COMP European Commission Expert Group for Rare Diseases European Parliament MEP Group Council of Europe and EDQM Comprehensive newsletters, 3 x year World Haemophilia Day events 37

The Future when I was Born: Peanuts for haemophilia Boudreaux HB & Frampton VL Nature 185: 469-47 (196) 38

What People with Haemophilia want? To avoid or prevent bleeds and micro bleeds To treat bleeds effectively and with minimum disruption when they occur To be able to lead a normal life and engage in normal activities Treatment that is safe and effective Treatment that is convenient and easy to administer 39

What People with Haemophilia want? Oral treatment not currently feasible Subcutaneous treatment Less frequent intravenous infusion EHL CFCs Treatment that lasts longer and gives higher factor levels or more protection Treatment designed with patients in mind as end users- not clinicians or nurses To go from a severe form of hemophilia (<1-2%) to a mild form (>5%) Possibility of a cure- Gene therapy 4

Ideal Treatment Options Non intravenous treatment Safe and effective Better treatment options for inhibitors More treatment options for VWD and RBDs Treatment that minimizes disruption of life Economically sustainable to allow access to treatments by payers 41

Agenda Welcome Josh Brodsky, Associate Director, Investor Relations & Corporate Communications Introduction Akin Akinc, Ph.D., Vice President, General Manager, Fitusiran Overview of Hemophilia and Patient Perspective Brian O Mahony, Chief Executive, Irish Haemophilia Society Ltd. and person living with severe hemophilia B Fitusiran Program Akin Akinc, Ph.D., Vice President, General Manager, Fitusiran Benny Sorensen, M.D., Ph.D., Senior Director, Clinical Research Q&A Session 42

Hemophilia and Rare Bleeding Disorders (RBD) Unmet Need Hemophilias are recessive X-linked monogenic bleeding disorders Hemophilia A: deficiency in Factor VIII (~16, diagnosed pts WW) Hemophilia B: deficiency in Factor IX (~4, diagnosed pts WW) Debilitating disease characterized by repeated bleeding into musculoskeletal system Very high treatment burden for prophylactic therapy Complications arise from repeated IV access Real-world efficacy suboptimal due to reduced compliance Inhibitor patients 1,2 represent a segment of very high unmet need 2, Patients in major markets; up to 6, WW >15-25 Bleeds/year; >5 in-hospital days/year ~$3,/year avg. cost; up to >$1M/year Other RBD with segments of unmet need Inadequate prophylaxis options for RBD patients with severe bleeding phenotypes 43 1 WFH 212 Global Survey; 2 Antunes et al., Haemophilia. 2:65-72 (214)

Hemophilia Emerging Therapeutic Landscape Exciting new approaches for the management of hemophilia are in development Modified Factor Replacement Therapies Physiochemical engineering of factor replacements to extend half-life and reduce immunogenicity Gene Therapy Single administration of gene vector with potential to lessen disease severity by eliciting continuous production of factor TFPI Inhibitors Agents blocking TFPI, an inhibitor of coagulation, as a way to promote coagulation Bispecific Antibodies Emicizumab (ACE91) is a bispecific IgG antibody that binds FIXa and FX, thus mimicking the function of FVIII cofactors and promoting thrombin generation RNA Interference Therapeutics Fitusiran is a sirna targeting antithrombin in order to increase thrombin generation 44

Fitusiran Investigational RNAi Therapeutic for Treatment of Hemophilia Fitusiran (ALN-AT3) SC-administered small interfering RNA (sirna) therapeutic targeting antithrombin (AT) Non-biologic, chemically-synthesized, with targeting ligand to specifically deliver to liver site of AT synthesis Harnesses natural RNA interference (RNAi) mechanism for regulation of plasma AT levels Hemophilia A FVIII Hemophilia B FVIIIa FX FVIIa FVII Therapeutic hypothesis Hemophilia A and B are characterized by ineffective clot formation due to insufficient thrombin generation Fitusiran is designed to lower AT, with goal of promoting sufficient thrombin generation to restore hemostasis and prevent bleeding Observation of ameliorated bleeding phenotype in patients with co-inheritance of thrombophilic traits in hemophilia 1-4 Supported by pre-clinical data 5 and emerging Phase 1 clinical results 6 FIX FIXa FXa AT FVa Prothrombin Thrombin Fibrinogen Fibrin Blood clot FV 45 1 Kurnik K, et al. Haematologica. 92:982-985 (27); 2 Ettingshausen E, et al. Thromb Haemost. 85:218-22 (21); 3 Negrier C, et al. Blood. 81:69-695 (1993); 4 Shetty S, et al. Br J Haematol. 138:541-544 (27); 5 Seghal A, et al. Nat Med. 21:492-497 (215); 6 Pasi J, et al. ASH. (215)

Fitusiran A New Approach Several emerging features for potential differentiation Not a biologic it is a manufactured RNAi therapeutic No reconstitution or mixing required Administered subcutaneously (SC), as opposed to intravenously (IV) Long duration of action, currently being evaluated in Phase1/2 studies, administered as once-monthly dose Not a factor, therefore may help avoid inhibitor formation due to reduced factor exposure and intensity Mechanism directed at addressing thrombin production defect, amenable to hemophilia A and hemophilia B, including those who have developed inhibitors and potentially other RBD Stable at room temperature no cold chain required 46

Agenda Welcome Josh Brodsky, Associate Director, Investor Relations & Corporate Communications Introduction Akin Akinc, Ph.D., Vice President, General Manager, Fitusiran Overview of Hemophilia and Patient Perspective Brian O Mahony, Chief Executive, Irish Haemophilia Society Ltd. and person living with severe hemophilia B Fitusiran Program Akin Akinc, Ph.D., Vice President, General Manager, Fitusiran Benny Sorensen, M.D., Ph.D., Senior Director, Clinical Research Q&A Session 47

Fitusiran Phase 1 Study Dose-Escalation Study in Four Parts Part A: Single-Ascending Dose (SAD) Randomized 3:1, Single-blind, Placebo-controlled, Healthy volunteers 3 mcg/kg x 1 SC, N=4 Part B: Multiple-Ascending Dose (MAD) Weekly dosing Open-label, Patients with Hemophilia A or B 15 mcg/kg qw x 3 SC, N=3 45 mcg/kg qw x 3 SC, N=6 75 mcg/kg qw x 3 SC, N=3 Part C: MAD Monthly dosing Open-label, Patients with Hemophilia A or B 225 mcg/kg qm x 3 SC, N=3 45 mcg/kg qm x 3 SC, N=3 9 mcg/kg qm x 3 SC, N=3 18 mcg/kg qm x 3 SC, N=3 8 mg qm x 3 SC, N=6 Part D: MAD Monthly dosing Open-label, Patients with Hemophilia A or B with inhibitors 5 mg qm x 3 SC, N=6 8 mg qm x 3 SC, N=6 Ongoing 48 5 patients participating in Part C previously participated in Part B qw, weekly; qm, monthly; SC, subcutaneous

Interim Fitusiran Phase 1 Study Results* Demographics & Baseline Characteristics, Parts B, C & D Part B Subcutaneous Weekly 3 Part C Subcutaneous Monthly 3 Part D Subcutaneous Monthly 3 15 mcg/kg N=3 45 mcg/kg N=6 75 mcg/kg N=3 225 mcg/kg N=3 45 mcg/kg N=3 9 mcg/kg N=3 18 mcg/kg N=3 8 mg N=6 5 mg N=6 Age, mean (SD) 28 (9) 42 (14) 4 (4) 37 (21) 37 (15) 38 (16) 46 (12) 32 (12) 33 (7) Hemophilia A Hemophilia B 2 1 6 2 1 2 1 2 1 3 3 3 3 5 1 Severe Moderate 3 6 3 2 1 3 2 1 3 5 1 6 Weight (kg), mean (SD) 76 (1) 8 (22) 82 (8) 85 (12) 76 (16) 76 (2) 71 (12) 74 (1) 73 (17) 49 Pasi et al., WFH, July 216

Interim Fitusiran Phase 1 Study Results* Safety/Tolerability, Parts B, C, & D No SAEs related to study drug Majority of AEs mild or moderate in severity AEs (excluding ISRs) in 1% of patients Upper respiratory tract infection (1%), arthralgia (1%) 11 (35%) patients reported drug-related ISRs, all mild Mostly pain and/or erythema at injection site One patient discontinued due to AE of non-cardiac chest pain; considered severe and possibly related Associated with transient elevations of ALT (1x ULN), AST (8x ULN), CRP and D-dimer; no increase in total bilirubin Extensive evaluation unremarkable; VTE excluded by serial CT angiograms and liver and lower extremity ultrasound This event resolved with symptomatic management, including antacids and analgesics No thromboembolic events or laboratory evidence of pathologic clot formation (D-dimer, platelet count, fibrinogen, and/or PT/INR) With exception of case noted above, no other clinically significant drug-related changes in laboratory parameters No instances of anti-drug antibody (ADA) formation Bleed events successfully managed with infusion of standard replacement factor or bypass agents 5 *Data transfer up to 11July216 Pasi et al., WFH, July 216 Adverse event grouping based on MedDRA-coded terms, excluding bleed events

% Mean (+/- SEM) AT Activity Relative to Baseline Interim Fitusiran Phase 1 Study Results* AT Lowering, Part C AT lowering after monthly dosing in patients with hemophilia A and B 125 Cohort 1 225 mcg/kg (N=3) Cohort 2 45 mcg/kg (N=3) Cohort 3 9 mcg/kg (N=3) Cohort 4 18 mcg/kg (N=3) Cohort 5 8 mg (N=6) 1 75 5 25 2 4 6 8 1 12 14 16 18 2 22 Days since first dose Onset Observation 51 *Data transfer: 3Jun216 Pasi et al., WFH, July 216

Relative Nadir AT Level (Mean +/- SEM, %) Interim Fitusiran Phase 1 Study Results* AT Lowering, Parts A, B & C Dose-dependent AT lowering Mean maximal AT lowering of 87 ± 1% at 8 mg fixed dose 1 9 8 7 6 5 4 3 2 1 3 mcg/kg (n=3) 15 mcg/kg (n=3) 45 mcg/kg (n=6) 75 mcg/kg (n=3) 225 mcg/kg (n=3) 45 mcg/kg (n=3) 9 mcg/kg (n=3) 18 mcg/kg (n=3) 8 mg (n=6) Part A (Single dose) Part B (Weekly dose) Part C (Monthly dose) 52 *Data transfer: 3Jun216 Pasi et al., WFH, July 216

Peak Thrombin Generation (nm) Interim Fitusiran Phase 1 Study Results* Thrombin Generation, Part B & C Post hoc analysis of thrombin generation by AT lowering quartiles Mean thrombin generation increase of 289% relative to baseline at AT lowering >75% (p<.1 ) 25 Boxes denote median and interquartile range 2 15 1 5 53 N=4 Healthy Volunteers AT Lowering < 25% (N=3) AT Lowering 25-5% (N=25) AT Lowering 5-75% (N=25) Patients with Hemophilia *Data transfer: 3Jun216; Pasi et al., WFH, July 216 %Change in Peak TG: p<.1 by Mann-Whitney test, when compared with AT3 lowering than <25% group AT Lowering >75% (N=16)

AT Activity (%) AT Activity (%) AT Activity (%) 14 Interim Fitusiran Phase 1 Study Results* Exploratory Analysis of Factor Equivalence Fitusiran achieved peak thrombin generation values equivalent to >4% Factor VIII Pre-dose factor administration used to establish individualized factor-peak thrombin relationship (in all 3 patients with pre-dose factor data) Plasma collected at -.5, 1, 2, 8, 24, and 48 hours post factor administration Samples analyzed for FVIII level and thrombin generation Peak thrombin achieved post fitusiran dose compared to peak thrombin achieved with >4% FVIII C1-1 (225 mcg/kg qm) 16 14 C4-3 (18 mcg/kg qm) 16 14 C5-6 (8 mg qm) 16 12 14 12 14 12 14 1 8 6 4 2 4% factor equivalence 12 1 8 6 4 2 Peak Thrombin (nm) 1 8 6 4 2 4% factor equivalence 12 1 8 6 4 2 Peak Thrombin (nm) 1 8 6 4 2 4% factor equivalence 12 1 8 6 4 2 Peak Thrombin (nm) -3 3 6 9 12 Days -3 3 6 9 12 Days -3 3 6 9 12 Days Pre-dose Factor Peak Thrombin AT % Lowering Peak Thrombin 54 *Data transfer: 3Jun216 Pasi et al., WFH, July 216

ABR Estimate, Mean (SEM) Interim Fitusiran Phase 1 Study Results* Exploratory Analysis of Bleed Events, Parts B & C Post hoc analysis of bleed events by AT lowering quartiles Includes more than 11 cumulative days with AT lowering >75% in 16 patients 3 2 1 55 AT Lowering <25% AT Lowering 25-5% AT Lowering 5-75% AT Lowering >75% Patients 3 27 25 16 Cumulative Days 733 1119 123 1128 Cumulative Bleeds 47 4 36 11 ABR, Mean (SEM)** 22 ± 5 15 ± 6 11 ± 3 5 ± 2 ABR, Median 1 6 1 *Data transfer: 3Jun216; Pasi et al., WFH, July 216 ABR, annualized bleeding rate; SEM, standard error of the mean Number of patients with time spent in quartile; For each patient, the ABR in each quartile is calculated by 365.24*(number of bleed events/number of days in quartile) **P-value <.5

% AT Lowering Interim Fitusiran Phase 1 Study Results* Exploratory Analysis of Bleed Events Post hoc analysis of bleed events during Onset and Observation periods Prospectively collected bleed events during Onset (Day -28) and Observation periods (Day 29 to last available, to maximum of Day 112) -2 2 4 6 AT % Lowering Peak Thrombin Bleed Event Factor Administration 16 14 12 1 8 6 4 Peak Thrombin (nm) 8 2 1 14 28 42 56 7 84 98 112 126 154 182 21 Time (Days) Onset Observation 56 *Data transfer: 3Jun216; ; Pasi et al., WFH, July 216 Patient has severe hemophilia A and has a self-reported ABR of 22; enrolled in Part B (45 mcg/kg dose cohort) Plot updated on 11Aug216 to reflect corrected peak thrombin values (reruns conducted on samples with assay errors)

Interim Fitusiran Phase 1 Study Results* Exploratory Analysis of Bleed Events, Part C Dose Patient Prior Tx 225 mcg/kg 45 mcg/kg 9 mcg/kg 18 mcg/kg 8 mg^ Pre-study ABR Onset ABR All Bleeds, n Observation Period ABR Spontaneous Bleeds, n C1-1 PPx 2 13 1 4 1 4 C1-2 PPx 1 4 C1-3 PPx 5 4 17 C2-1 PPx 4 25 4 17 3 13 C2-2 OD 38 13 C2-3 PPx 4 1 4 C3-1 PPx C3-2 OD 2 25 3 13 C3-3 OD 32 25 C4-1 PPx 25 C4-2 OD 24 C4-3 PPx 25 C5-1 PPx 12 13 2 9 1 4 C5-2 PPx 16 C5-3 PPx 6 13 2 9 C5-5 PPx 6 13 C5-6 PPx 13 AsBR 57 *Data transfer: 3Jun216; Pasi et al., WFH, July 216 PPx: Prophylaxis, OD: On-Demand; ABR, annualized bleeding rate; AsBR, annualized spontaneous bleeding rate Post hoc analysis of treated bleed events during Onset (Day -28) and Observation periods (Day 29 to last study visit or last dose+56 days, whichever is earlier; Pre-study ABR derived from medical records; ^Patient C5-4 withdrawn, excluded from analysis

ABR ABR Interim Fitusiran Phase 1 Study Results* Exploratory Analysis of Bleed Events, Part C Summary of Median ABR (All Cohorts, n=17^) Summary of Median ABR (8 mg, n=5) 3 25 28 15 13 2 1 15 13 6 1 5 5 2 PPx OD Onset Observation (n=13) (n=4) Pre-study Onset (n=17) Observation (n=17) Pre-Study PPx Onset Observation Pre-Study Pre-study Onset Observation All Part C patients^ Median ABR, Pre-study period: 2 (PPx); 28 (OD) Median ABR, Observation period: 53% of patients report no bleeds 82% of patients report no spontaneous bleeds Part C, 8 mg dosing cohort ^ Median ABR, Pre-Study (all PPx patients): 6 Median ABR, Observation period: 58 *Data transfer: 3Jun216; Pasi et al., WFH, July 216 PPx: Prophylaxis, OD: On-Demand; ABR, annualized bleeding rate; Post hoc analysis of treated bleed events during Onset (Day -28) and Observation periods (Day 29 to last study visit or last dose+56 days, whichever is earlier; Pre-study ABR derived from medical records; ^Patient C5-4 withdrawn, excluded from analysis

Fitusiran Phase 1 Study Inhibitor Patients, Part D Study population Hemophilia A and Hemophilia B patients with inhibitors, utilizing bypassing agents (BPAs) for bleed management Exploratory pre-dose evaluation of response to BPAs BPA administered prior to fitusiran dosing to explore peak thrombin response to patient s standard BPA Plasma collected at -1, 2, 6, and 24 hours post BPA administration, and samples analyzed for thrombin generation Fitusiran dose cohorts First cohort (N=6) enrolled and dosed at fixed, low monthly dose of 5 mg Second cohort (N=6) enrolled and being dosed at fixed, monthly dose of 8 mg Follow-up ongoing 59

AT Activity (%) AT Activity (%) AT Activity (%) AT Activity (%) AT Activity (%) AT Activity (%) Interim Fitusiran Phase 1 Study Results* AT, Peak Thrombin, Part D (Cohort 1, 5 mg) Initial AT lowering and thrombin generation results in patients with inhibitors Comparable AT lowering and thrombin generation as observed with similar doses in non-inhibitor patients Thrombin generation with fitusiran consistently exceeds transient levels achieved with BPA administration 14 12 1 8 6 4 2 D1-1 D1-2 D1-3 -3 3 6 9 12 Days 1 8 6 4 2 Peak Thrombin (nm) 14 12 1 8 6 4 2-3 3 6 9 12 Days 1 8 6 4 2 Peak Thrombin (nm) 14 12 1 8 6 4 2-3 3 6 9 12 Days 1 8 6 4 2 Peak Thrombin (nm) 14 D1-4 D1-5 D1-6 1 14 1 14 1 6 12 1 8 6 4 2-3 3 6 9 12 Days *Data transfer: 11July216 Pasi et al., WFH, July 216 8 6 4 2 Peak Thrombin (nm) 12 1 8 6 4 2 Pre-dose BPA Peak Thrombin -3 3 6 9 12 Days AT % Lowering 8 6 4 2 Peak Thrombin (nm) Peak Thrombin 12 1 8 6 4 2-3 3 6 9 12 Days 8 6 4 2 Peak Thrombin (nm)

Interim Fitusiran Phase 1 Study Results* Exploratory Analysis of Bleed Events, Part D (Cohort 1, 5 mg) Observation Period Dose Patient Hemophilia Prior Tx Prescribed BPA Pre-study ABR Onset ABR Days in Obs Period Bleeds, n ABR D1-1 HA w/ inh OD apcc 4 13 57 D1-2 HB w/ inh OD rfviia/apcc 26 13 55 2 13 5 mg D1-3 HA w/ inh OD rfviia ** N/A N/A D1-4 HA w/ inh OD apcc 52 38 56 4 26 D1-5 HA w/ inh OD PCC 8 38 55 6 4 D1-6 HA w/ inh OD rfviia 16 13 57 Data suggest partial effect at 5 mg (49-1% reduction in Pre-study ABR) Further follow-up ongoing to explore safety and bleed efficacy with longer-term dosing; all eligible patients (reaching Day 84) have rolled over to extension study Second cohort dosed at 8 mg; follow-up ongoing 61 *Data transfer: 11Jul216 Pasi et al., WFH, July 216 w/ inh, with inhibitors; PPx, Prophylaxis; OD, On-Demand; ABR, annualized bleeding rate **As of data transfer date, patient does not have sufficient follow up in Observation period Post hoc analysis of treated bleed events during Onset (Day -28) and Observation periods (Day 29 to last study visit or last dose+56 days, whichever is earlier); Pre-study ABR derived from medical records

Fitusiran Phase 1 Study* Summary of Interim Results Fitusiran generally well tolerated in hemophilia A and B patients with and without inhibitors No SAEs related to study drug; no thromboembolic events AEs (excluding ISRs) in 1% of patients: upper respiratory tract infection (1%) and arthralgia (1%); majority mild or moderate in severity 11 (35%) patients reported mild drug-related ISRs Mostly pain and/or erythema at the injection site 1 discontinuation due to AE; event resolved in this patient with symptomatic management Evidence of clinical activity and potential correction of hemophilia phenotype in noninhibitor patients Dose-dependent AT lowering and thrombin generation increase achieved, with once-monthly subcutaneous dose regimen; fixed 8 mg dose provides consistent AT lowering >75% In exploratory post-hoc analysis in monthly dose cohorts, fitusiran achieved median ABR =, with 53% patients bleed-free and 82% patients experiencing zero spontaneous bleeds Encouraging early data in inhibitor patients AT lowering and thrombin generation increase consistent with non-inhibitor patients Thrombin generation increases consistently exceed those achieved transiently with BPA administration Exploratory post-hoc analysis shows 49-1% reduction of bleeds at initial 5 mg dose Second cohort (N=6) now fully enrolled at 8 mg 62 *Data transfer: up to 11Jul216 Pasi et al., WFH, July 216

Fitusiran Program Next Steps 21 patients enrolled (as of July 11, 216) in open-label extension (OLE) study, including inhibitor patients As of the July 11, 216 data cutoff date, patients had received up to 13 monthly doses of fitusiran Additional data expected to be shared in late 216, likely at ASH, pending abstract approval Updated results from Parts C and D of the ongoing Phase 1 study Highest and fixed dose cohorts, including inhibitor patients Initial results from Phase 1/2 OLE study Plan to advance to pivotal studies in early 217 63

2:1 RANDOMIZATION 2:1 RANDOMIZATION Preliminary Fitusiran Phase 3 Design* Both studies expected to begin in early 217 STUDY 1 Population: Adults and adolescents with Severe Hemophilia A or B with inhibitors N~5 Fitusiran OR SOC Endpoints (at 9 months): Annualized Bleed Rate Total number of bleeds By-passing agent consumption QoL Safety STUDY 2 Population: Adults and adolescents with Severe Hemophilia A or B N~1 Fitusiran OR SOC Endpoints (at 9 months): Annualized Bleed Rate Total number of bleeds Factor VIII/IX consumption QoL Safety 64 *Preliminary plans subject to further diligence and health authority feedback Patients in both Study 1&2 will be allowed to roll over into open-label extension

Agenda Welcome Josh Brodsky, Associate Director, Investor Relations & Corporate Communications Introduction Akin Akinc, Ph.D., Vice President, General Manager, Fitusiran Overview of Hemophilia and Patient Perspective Brian O Mahony, Chief Executive, Irish Haemophilia Society Ltd. and person living with severe hemophilia B Fitusiran Program Akin Akinc, Ph.D., Vice President, General Manager, Fitusiran Benny Sorensen, M.D., Ph.D., Senior Director, Clinical Research Q&A Session 65

Upcoming RNAi Roundtables ALN-CC5 for the treatment of Complement-Mediated Diseases Wednesday, August 31, 11: a.m. 12: p.m. ET Jeff Miller, Vice President, General Manager, CC5 Program Pushkal Garg, M.D., Senior Vice President, Clinical Development Guest Speaker: Anita Hill, M.D., Ph.D., MRCP, FRCPath, Consultant Haematologist for Leeds Teaching Hospitals NHS Trust, UK, and Lead for the National PNH Service in England ALN-AS1 for the treatment of Acute Hepatic Porphyrias Tuesday, September 13, 11:3 a.m. 12:45 p.m. ET John Maraganore, Ph.D., Chief Executive Officer William Querbes, Ph.D., Associate Director, Research Guest Speaker: Ariel Lager, living with Acute Intermittent Porphyria ALN-GO1 for the treatment of Primary Hyperoxaluria Type 1 (PH1) Tuesday, September 27, 1: a.m. 11: a.m. ET Barry Greene, President and Chief Operating Officer David Erbe, Ph.D., Director, Research Guest Speaker: Sally-Anne Hulton, M.D., FRCPCH, MRCP, FCP, MBBCh, Consultant Paediatric Nephrologist and Clinical Lead, Birmingham Children s Hospital NHS Trust Guest Speaker: Jennifer Lawrence, M.D. (mother of George Tidmore, a PH1 patient) ALN-HBV for the treatment of Hepatitis B Virus (HBV) Infection Tuesday, October 11, 9: a.m. 1: a.m. ET Barry Greene, President and Chief Operating Officer Laura Sepp-Lorenzino, Ph.D., Vice President, Entrepreneur-in-Residence Guest Speaker: Heiner Wedemeyer, M.D., Managing Senior Physician and Assistant Professor in the Department of Gastroenterology, Hepatology and Endocrinology at Hannover Medical School For more information, please visit www.alnylam.com/roundtables 66

Thank you www.alnylam.com 67