* Your assessment is very important for improving the work of artificial intelligence, which forms the content of this project
Download n = 40 - Corporate
Survey
Document related concepts
Transcript
A Broad Clinical Pipeline NASDAQ: RIGL Forward Looking Statements Statements included in this presentation that are not historical in nature are “forward-looking statements” within the meaning of the “safe harbor” provisions of the Private Securities Litigation Reform Act of 1995. These statements include those regarding clinical trial plans, business strategy and research and development efforts. Forward-looking statements are subject to certain risk and uncertainties that could cause actual results to differ materially from those indicated in the forward-looking statements due to the risk factors identified in Rigel’s Form 10-Q for the quarter ended September 30, 2005 and other documents filed with the Securities and Exchange Commission. The information in this presentation is current as of this date. Rigel takes no responsibility to update the information. Today’s Agenda Introduction James M. Gower Rheumatoid Arthritis: Dr. Lee S. Simon Current Therapies And Regulatory Review Rigel Product Pipeline Questions and Answers Dr. Donald G. Payan Investment Thesis: A Broad Pipeline of Major Opportunities Clinical Milestones Next 12 Months R788 in Rheum. Arthritis Phase II trial start 1H, 2006 R788 in ITP/Flt3 AML Phase I/II trials start 2006 R763 in Oncology Phase I/II trial start 1H, 2006 Rxxx in Transplant Rejection Preclinical Development 2006 Ryyy in Asthma Preclinical Development 2006 Angiogenesis/Cancer Select Clinical Candidate, 2006 Our Business Strategy: Many Ways to Win Portfolio strategy for drug development > Multiple novel products to reduce risk Develop or co-develop by product Partnering after proof of human efficacy provides greater value Focus primarily on large markets Focus programs on Immunology and Infectious Disease > Secondary focus on oncology Small molecule therapeutics A History of Successful Large Pharma Collaborations >$140M from collaborations to date Focused on immunology and oncology Complements internal research Early collaborations all expanded/extended Asthma/COPD ‘05 Immune Diseases ‘99 Oncology ‘98 Angiogenesis ‘01 Immune Diseases ‘99 Ligase Targets in Oncology ‘04 Ligase Target in Oncology ‘02 Aurora Kinase Inhibition in Oncology ‘05 Key Financial Highlights $127.9M total cash at end Q3, 2005 $25.0M additional cash with Serono partnership in Q4, 2005 $12M - $14M net cash burn per quarter Sufficient cash to move products to later stages of development Incremental cash from future milestones and new partnerships 24.8 million shares outstanding, 28.7 million fully diluted Lee S. Simon, M.D. Associate Clinical Professor, Harvard Medical School Associate Clinical Professor at Harvard Medical School and head regulatory consultant with MEDACorp, a management consulting group that offers strategic advice to leading life sciences and healthcare companies Previously, Division Director of the Arthritis, Analgesic and Ophthalmologic Drug Product Division at the FDA Prior to his tenure at the FDA, Director of Rheumatology Clinical Research and Associate Chief of Medicine at Beth Israel Deaconess Medical Center in Boston, MA Medical Need for Novel Therapeutic Options in the Treatment of Rheumatoid Arthritis Lee S. Simon, MD Associate Clinical Professor of Medicine Harvard Medical School Beth Israel Deaconess Medical Center Rheumatoid Arthritis • Affects about 1% of the US population between the ages of 20-50 • Heterogeneous disease • Variable course • Systemic inflammatory disease associated with an as yet poorly understood immune dysfunction • Leads to the development of destructive erosive disease in a great majority • Treatment to date has been to alter the inflammatory cascade to lead to decreased inflammation and decrease in pain • Remission rare, cure not yet observed Rheumatoid Arthritis • Shortens life span • Untreated: increased risk for non-Hodgkins lymphoma • Most patients suffer an unrelenting course characterized by recurrent flares over years leading to progressive loss of functional status and ultimately leading to significant disability; an unfortunate few have an accelerated mutilating course and a lucky few have either mild disease or enter into remission early RA Is a Progressive Disease Early Intermediate Late Severity Inflammation Disability Radiographs © ACR 0 5 10 15 20 Duration of Disease (y) Graph: Adapted from Kirwan JR. J Rheumatol. 2001;28:881-886. Photo: © American College of Rheumatology. 25 30 Disability in RA with Standard of Care Percent Maximum Disability 70 60 50 Wolfe et al Lassere et al Sherrer et al Scott et al 40 30 20 10 0 7 12 Disease Duration (years) Scott DL et al. Rheumatol. 2000; 39:122-132. 18 Joint Erosions Occur Early in RA Up to 93% of patients with < 2 years of RA may have radiographic abnormalities • Erosions can be detected by MRI within 4 months of RA onset • Rate of progression is significantly more rapid in the first year than in the second and third years Maximum percent of joints affected • MTP 30 Total Hand 20 10 0 0 1 Year MRI, magnetic resonance imaging; MTP, metatarsophalangeal. Fuchs HA et al. J Rheumatol. 1989;16:585-591; McQueen FM et al. Ann Rheum Dis. 1998;57:350-356; van der Heijde DM et al. J Rheumatol. 1995;22:1792-1796. 2 3 Rheumatoid Arthritis • Chronic inflammatory autoimmune disease – Involves synovial membrane and extra-articular sites – Associated with rheumatoid factor (autoantibody) production • Genetic predisposition – Familial incidence – Genetic factor: HLA-DR4-related antigens • Unknown environmental trigger Impact of RA on Health-Related Quality of Life • • • • • • Pain and suffering Decreased physical functioning Increased psychological distress Decreased social functioning Increased healthcare utilization Increased work disability Treatment Goals for Arthritis Patients • • • • • • • Halt progression of disease Maximize functional independence Optimize treatment of pain Optimize treatment of inflammation Enhance health related quality of life Minimize potential for toxicity Provide easy access to care at reasonable cost Arthritis Management, 1892 “Many cases are greatly helped by prolonged residence in southern Europe or southern California. Rich patients should always be encouraged to winter in the south and in this way avoid cold, damp weather.” Osler, Principles and Practice of Medicine, 1892 Evolution of RA Treatment MTX, Oral gold Injectable gold HCQ, Steroids 1930s B-cell antagonists, CTLA-4 Ig, IL-6 receptor antagonist 1950s SSZ 1960s D-Pen, AZA 1970s TNF antagonists, LEF, Cyclosporine 1980s 1990s Remission/ Cure 2000s Early DMARD treatment Treat signs and symptoms in established disease Aggressive MTX dosing Combination therapy AZA, azathiopine; Ig, immunoglobulin; HCQ, hydroxychloroquine; LEF, leflunomide; SSZ, sulfasalazine. FDA Approval RA Threshold for US • Signs and symptoms – Typical show ACR 20 response is better than placebo • Improvement in function – HrQol improvement over placebo was 2 years as of March 5, 2003 only 1 year data required • Altering X ray progression – One year data required with second year for durability of response • Major clinical response – An ACR 70 response on drug for 6 months to 1 year • Remission – No evidence of active disease off all drugs for 6 months What We Have Achieved • We have drugs that as monotherapy provide benefit of improvement in signs and symptoms in about 50% of patients • Combination therapy at times leads to improvements approaching 75% • We have drugs that appear to delay x ray progression which if patients respond include all DMARTs • We do not cure this disease as of this time ACR 20 • Tender joint count • Swollen joint count • • • • • VAS scale for pain Patient global Physician global HAQ-DI ESR or CRP ACR 20 Components That are Inter-related • • • • • Pain Patient global Physician global HAQ-DI Tender joint counts Current Drug Therapy Options in Rheumatoid Arthritis • Non-selective NSAIDs (Rx, OTC)/ Selective COX-2 Inhibitors • Disease Modifying Anti-rheumatic Drugs (DMARDs) • Immunosuppressives • Glucocorticoids • Biologic agents • Investigational agents Drug Therapy • For many years it was considered standard of care to be cautious and not expose patients to potentially toxic therapy which had not clearly been shown to alter disease • Diagnosis was faulty and many early patients suffered probably viral arthritis and not true RA; many early spontaneous remissions were likely due to a viral etiology • Thus a treatment “pyramid” emphasized slow progression of therapy from least effective modalities to palliate pain and suffering to potentially more effective but also those therapies associated with more risk of adverse events The New Treatment Paradigm Celecoxib vs Naproxen in RA Number of Tender/Painful Joints at Week 12 00 Placebo Placebo (n (n == 231) 231) Mean Change Change Mean From Baseline Baseline From Fewer -2 -2 Celecoxib Celecoxib 100 100 mg mg BID BID (n (n == 240) 240) -4 -4 -6 -6 -8 -8 -10 -10 -12 -12 -14 -14 --8.2 8.2 --10.1 10.1 --12.0 12.0 --12.3 12.3 ** ** Celecoxib Celecoxib 200 200 mg mg BID BID (n (n == 235) 235) Naproxen Naproxen 500 500 mg mg BID BID (n (n == 225) 225) Mean Mean baseline baseline values: values: placebo placebo == 28.7; 28.7; celecoxib celecoxib 100 100 mg mg BID BID == 29.6; 29.6; celecoxib celecoxib 200 200 mg mg BID BID == 31.0; naproxen 500 mg BID = 28.3. 31.0; naproxen 500 mg BID = 28.3. *P *P << 0.05 0.05 vs vs placebo. placebo. Simon Simon LS, LS, et et al. al. JAMA. JAMA. 1999;282:1921–1928. 1999;282:1921–1928. Drug Therapy • Known “truths” – NSAIDs were/are palliative: do not alter fundamental disease – DMARDs • Important for those patients with progressive disease: likely would take 6 months to know benefit • Highly toxic, were associated with significant risk • Required weekly surveillance with initiation of therapy and if tolerated would still require monthly visits • Many patients did not have an adequate response • Standard of care was still associated with damage evident by x-ray and progressive loss of functional status Drugs Used to Treat RA Prior to 1985 • • • • • • • Antimalarials IM Gold Penicillamine Cyclosporins Azathioprine Cyclophosphamide Chlorambucil Drug Therapy • Antimalarials – Fortuitously discovered when given for either antimalarial prophylaxis or treatment in World War II to people with RA. Is not a DMARD • IM gold – previously used to treat some infections: may be a DMARD – 1966 Empire Rheumatism Council studied IM gold therapy demonstrating significant improvement and an occasional case of “remission” with significant risk in over 40% of patients: • Heavy metal induced kidney damage • Bone marrow suppresion • Liver effects, skin, vasculitis • Cyclophosphamide – Significant benefit with decreasing disease activity and x-ray benefit – Chronic oral therapy increased risk of urogenital cancer, leukemia, immunosuppression, bone marrow failure, nausea vomiting, hair loss Drug Therapy • Methotrexate – First studied at “low dose” in the 1960’s: concerns surrounded use of chemotherapy agent in chronic “non-fatal” disease – 1985 new description of use at 7.5 mgs weekly showing benefit – Subsequently more common dose is 15-25 mgs weekly – Better tolerated than previous DMARDS – Some evidence of true disease modification – Potential adverse effects included progressive liver damage even with consistent monitoring, lung fibrosis, acute pulmonary disease, bone marrow suppression, immunosuppression Estimated Continuation of Initial Secondline Therapy Over 60 Months Pincus et al. J Rheumatol 19:1885, 1992. Disease Modifying Anti-Rheumatic Therapies (DMARTs) • • • • Sulfasalazine Methotrexate Leflunomide Biologic response modifiers – TNF alpha inhibitors • Etanercept • Infliximab • Adalimumab – IL-1ra Potential Future Treatments • T-cell inhibitors • – abatacept • B-cell directed therapy – – – – • Anti-CD20 Anti-BLyS Anti-CD22 Anti-BAFF Other cytokine blockers – TNF-α–converting enzyme – IL-6 receptor antagonist Chemokine inhibitors – CCR2 inhibitors • Inhibitors of T-cell activation – CTLA-4 Ig – Rapamycin-like drugs • Intracellular signal blockade – P38 MAPK inhibitors – IKK inhibitors – Other kinase inhibitors BLyS, B lymphocyte stimulator; BAFF, B-cell activating factor; CTLA-4, cytotoxic T-lymphocyte-associated antigen 4; MAPK, mitogen-activated protein kinase. Shanahan JC et al. Curr Opin Rheumatol. 2003;15:226-236; Baker KP et al. Arthritis Rheum. 2003;48:3253-3265; Foey A et al. Arthritis Res. 2002;4:64-70; Kaufmann J et al. European League Against Rheumatism (EULAR) 2004; June 9-12, 2004; Berlin, Germany; Vaux DL. J Clin Invest. 2002;109:17-18. Advantages of DMARTs • Have been shown to: – Slow disease progression – Improve functional disability – Decrease pain – Interfere with inflammatory processes – Retard development of joint erosions Summary of ACR Response Rates* for Leflunomide, Sulfasalzine, Methotrexate ACR 20% ACR 50% ACR 70% 52.2 26.3 45.6 34.3 7.6 22.8 20.2 4.2 9.4 MN301(6 months) Leflunomide (n=130) Placebo (n=91) Sulfasalazine (n=132) 54.6 28.6 56.8 33.1 14.3 30.3 10.0 2.2 7.6 Non-PlaceboActiveControlled Studies MN302 (12 months) Leflunomide (n=495) Methotrexate (n=489) 51.1 65.2 31.1 43.8 9.9 16.4 Study and Treatment Group Placebo-Controlled Studies US301 (12 months) Leflunomide (n=178) Placebo (n=118) Methotrexate (n=180) * Intent to treat (ITT) analysis using last observation carried forward (LOCF) technique for patients who discontinued early. N is the number of ITT patients for whom adequate data were available to calculate the indicated rates. p<0.001 leflunomide vs placebo p<0.02 leflunomide vs placebo PERCENTAGE OF PATIENTS WHO ACHIEVED AN ACR RESPONSE AT WEEKS 30 AND 54 with infliximab REMICADE + MTX 3 mg/kg a 10 mg/kg a Placebo + MTX (n=88) q 8 wks (n=86) q 4 wks (n=86) q 8 wks (n=87) q 4 wks (n=81) Week 30 20% 50% 50% 52% 58% Week 54 17% 42% 48% 59% 59% ACR 50 Week 30 5% 27% 29% 31% 26% Week 54 9% 21% 34% 40% 38% ACR 70 Week 30 0% 8% 11% 18% 11% Week 54 2% 11% 18% 26% 19% Response ACR 20 a p < 0.05 for each outcome compared to placebo The ACR 20, 50 70 Responses with Etanercept Placebo Controlled Study I Study II Active Controlled Study III Placebo ENBRELa MTX/Placeb MTX/ENBRELa MTX o Response N = 80 N = 78 N = 30 N = 59 N = 217 ACR 20 Month 3 23% 62%b 33% 66%b 56% b b Month 6 11% 59% 27% 71% 58% Month 12 NA NA NA NA 65% ENBRELa N = 207 62% 65% 72% ACR 50 Month 3 Month 6 Month 12 8% 5% NA 41%b 40%b NA 0% 3% NA 42%b 39%b NA 24% 32% 43% 29% 40% 49% ACR 70 Month 3 Month 6 Month 12 4% 1% NA 15%b 15%b NA 0% 0% NA 15%b 15%b NA 7% 14% 22% 13%c 21%c 25% a.25 mg ENBREL SC twice weekly. b. p < 0.01, ENBREL vs. placebo. c. p < 0.05, ENBREL vs. MTX. ACR Responses in Placebo-Controlled Trials of Humira (adalimumab) (Percent of Patients) Response Study II Monotherapy (26 weeks) Placebo HUMIRA HUMIRA 40 mg 40 mg weekly every other N=103 week N=110 N=113 Study III Methotrexate Combination (24 and 52 weeks) Placebo/MT X HUMIRA/MTX 40 mg every other week N=200 N=207 ACR20 Month 6 Month 12 19% NA 46%* NA 53%* NA 30% 24% 63%* 59%* 8% NA 22%* NA 35%* NA 10% 10% 39%* 42%* 2% NA 12%* NA 18%* NA 3% 5% 21%* 23%* ACR50 Month 6 Month 12 ACR70 Month 6 Month 12 *p<0.01, HUMIRA vs. placebo Percent of Patients with ACR Responses of IL1-ra Response Study 1 (Patients on MTX) Study 3 (No DMARDs) TM Placebo Kineret TM Placebo Kineret 100 mg/day 75 mg/day 150mg/day (n=251) (n=250) (n=119) (n=115) (n=115) ACR 20 Month 3 Month 6 24% 22% 34% a 38% c ACR 50 Month 3 Month 6 6% 8% 13% b 17% b 5% 8% 10% 11% 8% 19%a ACR 70 Month 3 Month 6 0% 2% 3% a 6% a 0% 1% 0% 1% 0% 1% a b p<0.05, KineretTM versus placebo p<0.01, KineretTM versus placebo c 23% 27% p<0.001, KineretTM versus placebo 33% 34% 33% 43%a LEF: Mean Improvement in HAQ DI Over Time Year-2 Cohorts: 0–24 Months Improved HAQ DI 1.7 1.5 US301 (n = 97) MN301/3/5 (n = 51) MN302/4 (n = 248) 1.3 1.1 32% 46% 0.9 0.7 50% 0.5 0.3 Baseline 6 mo Strand V, et al. J Rheumatol. 2005; in press. 12 mo 24 mo Etanercept: Mean Change in HAQ DI Over 2 Years (ERA) 1.7 Methotrexate Etanercept (25 mg) Mean HAQ DI Improved 1.5 1.3 1.1 0.9 0.8 0.7 0.5 0.3 Baseline Month 6 Kosinski M et al. AJMC. 2002;8,:231-240 Month 12 Month 18 Month 24 INF: Mean HAQ DI Over Time Year-2 Patients 0–24 Months (ATTRACT) 1.7 1.7 HAQ DI 1.5 1.3 1.5 1.5 1.5 1.3 1.3 1.3 1.6 1.3 1.1 Improved 0.9 All INF + MTX 0.7 0.5 ITT Completed 54 wk Baseline 6 mo (n = 340) (n = 296) Placebo + MTX (n = 86) (n = 50) 12 mo Maini RN, et al. Arthritis Rheum. 2004;50:1051–1065; Kavanaugh AF, et al. Arthritis Rheum. 2000;43:S147. 24 mo Adalimumab+MTX: Mean Change in HAQ DI over 2 years: DE019 All adalimumab treated patients 1.6 Excludes placebo-treated patients 1.4 1.2 1 0.8 0.6 0.4 0.2 At last visit, 31% of patients had HAQ DI scores of 0 0 0 6 Mos Keystone E, et al. EULAR 2004:FRI0098 12 Mos 18 Mos 24 Mos ASPIRE: MTX v MTX+INF in Early RA Demographics Median age (years) Median disease duration (years) Prior DMARD Rx (%) Corticosteroids (%) RF+ (%) HAQ DI [median] DAS28 at Baseline [median] Baseline Total Sharp Scores Erosion score ≤0 (%) MTX MTX+INF 3 mg/kg MTX+INF 6 mg/kg N=298 N=373 N=378 51 0.6 34 37 71 1.5 6.8 5.05 20 52 0.6 28 36 73 1.5 6.8 5.15 17 49 0.6 31 39 71 1.5 6.8 5.25 16 Smolen et al. Ann Rheum Ds 2003;62:S64 Intent to Treat Analysis Intent to Treat Analysis TEMPO: Trial of Etanercept and Methotrexate with Radiographic Patient Outcomes Design: Double-blind, randomized active controlled trial conducted in Europe and Australia Treatment: MTX vs ETN vs MTX+ETN over 52 weeks Patients: 682 patients with active RA and inadequate response to ≥ 1 DMARD; no Rx with MTX for ≥ 6 months 1° end point: AUC of ACR-N (24 weeks) Change in total Sharp score at 52 weeks 2° end points: ACR 20/50/70 responses DAS HAQ DI TEMPO: Demographics MTX N=228 Mean age (years) 53 Mean disease duration (years) 6.6 RF+ (%) 74 Mean DMARDs failed 2.3 Previous MTX use (%) 43 Corticosteroids (%) 57 Mean MTX dose (mg/wk) 17 Baseline DAS28 5.5 Baseline HAQ DI 1.7 -1.8 Baseline Total Sharp Scores 45 ETN N=223 MTX+ETN N=231 [<1 – 26 years] [0 – 9] Mean Changes in HAQ DI at Weeks 52 TEMPO RCT Baseline HAQ DI: 1.7 1.7 1.8 -0.1 Mean Change from BL -0.2 MCID -0.3 MTX -0.4 ETN -0.5 MTX+ETN -0.6 -0.7 -0.61 -0.66 -0.8 -0.9 -1.0 -0.97 Percentage of Patients Achieving and Not Achieving ACR Goals: TEMPO Trial* 90 Percent of Patients Achieving ACR Goals Percent of patients 80 Percent of Patients Not Achieving ACR Goals 70 60 50 40 30 20 10 0 ACR20 *Etanercept plus MTX arm. Klareskog L et al. Lancet. 2004;363:675-681. ACR50 ACR70 CTLA4Ig (abatacept) MOA • Activation of T cells requires 2 signals – Signal 1: antigen-specific interaction with the HMC-peptide complex – Signal 2: “costimulatory” interaction of specific receptors on T cells (CD28) with ligands on the APCs (CD80 and CD86) • • CTLA4Ig binds CD80 and CD86, which blocks the second signal In addition to blocking costimulation, CTLA4Ig may modulate the immune system through changes occurring in the APCs following the binding to cell surface ligands B7-1 (CD80) T Cell Activation B7-2 (CD86) T Cell CD28 Antigen Presenting Cell B7 (CD -1 80) B7 (CD -2 86) g CTLA4I T Cell An Pre tigen sen t Cel ing l CD28 By binding to CD80 and CD86, CTLA4Ig prevents interaction with CD28 on T-cells, preventing T-cell activation CTLA4Ig ACR 20, 50 & 70 Responses @ 6 Months Response Rate 80 ACR 20 ACR 50 ACR 70 Placebo + MTX CTLA4Ig 2mg/kg + MTX CTLA4Ig 10mg/kg + MTX ** 60 60 41.9 ** 36.5 35.3 40 20 * 22.9 * 10.5 11.8 1.7 0 * = p<0.05; ** = p<0.001 Treatment Groups ** 16.5 Rituximab Anti-CD20 Monoclonal Antibody • Chimeric murine/human monoclonal antibody – Variable light and heavy chain regions from murine model – Human IgG1, kappa constant region RTX • Long serum half-life (375 mg/m2) – Single dose t1/2 = 76 hours – Multidose t1/2 = 206 hours Berinstein NL et al. Ann Oncol. 1998;9:995-1001; Maloney DG et al. J Clin Oncol. 1997;15:3266-3274; Maloney DG et al. Blood. 1997;90:2188-2195. Rituximab: Mechanism of Action Antibody -Dependent Cell -Mediated Cytotoxicity Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC) (ADCC) Rituximab antibody Macrophage, monocyte, or natural killer cell CD20 Fcγ Fc RI, Fcγ Fc RII, or Fcγ Fc RIII Cell lysis Complement -Dependent Cytotoxicity Complement-Dependent Cytotoxicity (CDC) (CDC) B cell Complement activation (C1qC1rC1s) Membrane attack complex (MAC) CD20 Cell lysis Apoptosis Apoptosis Anderson DR et al. Biochem Soc Trans. 1997;25:705-708; Golay J et al. Blood. 2000;95:3900-3908; Reff ME et al. Blood. 1994;83:435-445; Clynes RA et al. Nat Med. 2000;6:443-446; Shan D et al. Cancer Immunol Immunother. 2000;48:673683; Silverman GJ et al. Arthritis Rheum. 2003;48:1484-1492. Rituximab in RA Phase IIa: Dosing Regimen and Treatment Schedule Rituximab 1 g IV or placebo Study day 1 Rituximab 1 g IV or placebo 3 8 15 CTX 750 mg IV or placebo 17 CTX 750 mg IV or placebo Methylprednisolone (100 mg IV) Prednisone (po) MTX or placebo weekly 60 mg/day Day 2, Days 4–7 30 mg/day Days 8–14 Continue to 24 weeks Preliminary data. Adapted from Szczepański L et al. European League Against Rheumatism (EULAR) 2003; June 18-21, 2003; Lisbon, Portugal. Rituximab in RA Phase IIa: ACR Responses at 24 Weeks (LOCF) Primary end point 76 † 80 73 ‡ MTX 65* 70 RTX RTX + CTX Percent 60 50 40 41 38 || 43 RTX + MTX || 33 § 30 23 20 15 13 15 5 10 0 ACR20 ACR50 ACR70 *P = 0.025, †P = 0.001, ‡P = 0.003, §P = 0.059, ||P = 0.005, ¶P = 0.048. P values using Fisher’s exact test, comparing MTX with each rituximab group. LOCF, last observation carried forward. Edwards JCW et al. N Engl J Med. 2004;350:2572-2581. ¶ Rituximab in RA Phase IIa: Improvement in Patient-Reported Outcomes (24 Weeks) MTX (n = 40) RTX (n = 40) RTX + CTX RTX + MTX (n = 41) (n = 40) Global disease activity (mm)* −14.0 −31.1 (P = 0.004) −31.0 −28.6 (P = 0.005) (P = 0.023) Pain (mm)* −13.3 −25.8 (P = 0.024) −25.2 −26.5 (P = 0.039) (P = 0.028) HAQ-DI* −0.3 −0.7 (P = 0.008) −0.5 (P = NS) −0.6 (P = 0.026) Preliminary data. *Mean change (negative values indicate improvement). HAQ-DI, Health Assessment Questionnaire Disability Index. Pavelka K et al. European League Against Rheumatism (EULAR) 2004; June 9-12, 2004; Berlin, Germany. Rituximab in RA Phase IIa: ACR Responses at 48 Weeks (NRI) 80 MTX 65* 70 RTX RTX + CTX Percent 60 50 49 40 30 20 RTX + MTX § 35 33 27 20 † § 15 5 10 10 0 0 ACR20 8 ACR50 *P < 0.0001, †P = 0.003, ‡P = 0.03, §P = 0.01. P values using Fisher’s exact test, comparing MTX with each rituximab group. NRI, Non-responder imputation. Edwards JCW et al. N Engl J Med. 2004;350:2572-2581. ACR70 15 ‡ Rituximab in RA Phase IIa: B-Cell Depletion and Recovery 4 Arms 3 Median CD19 (cells x 10 /µL) 500 MTX RTX RTX + CTX RTX + MTX 450 400 350 300 250 200 150 100 Lower limit of normal 50 0 0 4 8 12 16 20 24 28 32 36 40 44 48 Time (weeks) IV Steroids given on Days 1, 3, 15, and 17. Edwards JCW et al. N Engl J Med. 2004;350:2572-2581; Emery P et al. Arthritis Rheum. 2003;48:S439 (Preliminary data). Rituximab in RA Phase IIa: Most Frequently Reported Adverse Events (0–48 Weeks) All All events* events* 0–48 0–48 weeks weeks Most Most frequently frequently reported reported adverse adverse events events (≥ (≥ 5%) 5%) RA RA exacerbation exacerbation Hypotension** Hypotension** Hypertension** Hypertension** Nasopharyngitis Nasopharyngitis Arthralgia Arthralgia Back Back pain pain Hyperglycemia Hyperglycemia MTX (n = 40) 145 (85%) RTX (n = 40) RTX + CTX RTX + MTX (n = 41) (n = 40) 169 (88%) 161 (85%) 138 (85%) 55% 18% 15% 15% 8% 8% 40% 30% 18% 10% 8% 13% 37% 29% 7% 7% 5% 7% 18% 10% 5% 7% 8% 18% 25% 15% 13% 3% Cough Cough Flushing Flushing – 15% 5% 8% 8% 13% 5% 3% Headache Headache 5% 5% 7% 8% *Number of events reported by percentage of patients. **Hypotension/hypertension defined as > 30 mmHg change. Edwards JCW et al. N Engl J Med. 2004;350:2572-2581. Rituximab in RA Phase IIa: All Serious Infection Events (0–48 Weeks) MTX (n = 40) RTX (n = 40) RTX + CTX (n = 41) RTX + MTX (n = 40) Pneumonia (Pseudomonal) — — 1 — Bronchopneumonia* — 1 — — Septic Arthritis — 1 1 — Septicemia — — 1 — Preliminary data. All serious infection events (SIEs) occurred within the first 24 weeks. *Patient death. Szczepański L et al. 67th Annual Meeting of the American College of Rheumatology, October 23-26, 2003; Orlando, FL. Unanswered Questions About B-Cell Therapies • What is the role of steroids used with rituximab? • What is the duration of clinical efficacy after a single course of rituximab? • Is there a risk of increased occurrence of serious infections with B-cell targeted therapies? Opportunistic infections? • What role might biologics play in combination with B-cell targeted therapies? Unanswered Questions: Efficacy • Will the short- and long-term effects of different B-cell targeted therapies differ because of different mechanisms of action? • What is the duration of clinical efficacy after a single course of rituximab? • Will combination therapy (DMARDs + B-cell therapies) improve outcomes vs monotherapy? • Will combinations with biologics work as or more effectively? Unanswered Questions: Efficacy (cont’d) • What is the effect and duration of a second course of treatment? • Should a retreatment schedule be fixed or variable? • What are the effects on long-term radiographic damage and long-term disability? • What is the prevalence of human antichimeric antibodies and clinical/safety effects? Overview of Specific AEs with the Available DMARTs • • • • • Hepatic failure and hepatotoxicity Tuberculosis and other infections Interstitial lung disease Lymphoma Demyelinating disorders Hepatic Failure and Hepatotoxicity AE Reporting Rates for ETN, INF, LEF, and MTX Sept 1998–June 2003 Cumulative reporting rates per 100,000 pt-y (95% CI) AE Acute hepatic failure ETN INF 6.54 (3.94,10.22) 8.55 (6.19,11.2) LEF MTX 7.9 0.6 (5.69,10.67) (0.48,0.73) Denominators for exposure were estimated based on IMS data for the use of each agent for all indications. Pt-y of exposure calculated were 290,355 for ETN; 502,646 for INF; 531,959 for LEF; and 15,675,396 for MTX. Reported rates of AEs per 100,000 pt-y of drug exposure were calculated by dividing the number of AEs reported by pt-y of drug exposure. ETN = etanercept; INF = inflixamab; LEF = leflunomide; MTX = methotrexate. Cannon GW, et al. Presented at: Annual Meeting of the American College of Rheumatology (ACR); October 16–21, 2004; San Antonio, Tex. Presentation 1469. Tuberculosis and Other Infections AE Reporting Rates for ETN, INF, LEF, and MTX Sept 1998–June 2003 Cumulative Reporting Rates per 100,000 Pt-y (95% CI) AE ETN Sepsis/TB INF LEF 102.63 136.28 33.27 (91.31,114.97) (126.26,146.88) (28.55,38.55) MTX 4.87 (4.53,5.23) Drug exposure during observation period (100,00 pt-y) ETN – 290,355 INF – 502,646 LEF – 531,959 MTX – 15,675,396 Cannon GW, et al. Presented at: Annual Meeting of the ACR; October 16–21, 2004; San Antonio, Tex. Presentation 1469. Serious Infection Rates in Patients Receiving Biologic Therapy in the UK: BSRBR ETN 2602 2508 INF 2781 936 ADA 915 641 132 205 40 52.6 /1000 pt-yrs 52.1 /1000 pt-yrs 62.4 /1000 pt-yrs TB cases 2 9 0 Septic Joints 3 0 0 [n] patient yrs All Serious Infections Dixon WG, et al. EULAR 2005, #OP0094; Dixon WG, et al. ibid, #SAT0053; Dixon WG, et al. Rheumatol 2005;44(suppl 1):i11 Serious Infection Rates in Pts Receiving Biologics in the UK ETN Pt y ADA 936 641 132 (52.6) 205 (52.1) 40 (62.4) Lower respiratory 42 (16.7) 85 (21.6) 12 (18.7) Skin and soft tissue 26 (10.4) Bone and joint 15 (6.0) 21 (5.3) 3 (4.7) Urinary tract 12 (4.8) 13 (3.3) 6 (9.4) All serious infections 2508 INF 37(9.4) Entries presented as no. of infections (rate/1000 pt y). Dixon WG, et al. EULAR; June 8–11, 2005; Vienna, Austria. Abstract OP0094. [Evidence Level B] 8 (12.5) Interstitial Lung Disease (ILD) AE Reporting Rates for ETN, INF, LEF, and MTX Sept 1998–June 2003 Cumulative Reporting Rates per 100,000 Pt-Y (95% CI) AE ETN INF LEF MTX ILD 24.8 (19.4,31.23) 33.42 (28.56,38.88) 15.41 (12.26,19.13) 2.26 (2.03,2.51) Denominators for exposure were estimated based on IMS data for the use of each agent for all indications. Pt-y of exposure calculated were 290,355 pt-y for ETN; 502,646 pt-y for INF; 531,959 pt-y for LEF; and 15,675,396 pt-y for MTX. Reported rates of AEs per 100,000 pt-y of drug exposure were calculated by dividing the number of AEs reported by pt-y of drug exposure. Cannon GW, et al. Presented at: Annual Meeting of the ACR; October 16–21, 2004; San Antonio, Tex. Presentation 1469. Lymphoma AE Reporting Rates for ETN, INF, LEF, and MTX Sept 1998–June 2003 Cumulative Reporting Rates per 100,000 Pt-Y (95% CI) AE ETN Lymphoma INF LEF MTX 27.55 29.25 5.26 1.38 (21.85,34.29) (24.71,34.37) (3.50, 7.61) (1.21,1.58) Drug exposure during observation period (100,00 pt-y) ETN – 290,355 INF – 502,646 LEF – 531,959 MTX – 15,675,396 Cannon GW, et al. Presented at: Annual Meeting of the ACR; October 16–21, 2004; San Antonio, Tex. Presentation 1469. Demyelinating Disorders Demyelinating Reports*/100,000 Pt-Y Demyelinating Disorders: AERS Reporting Rates *FDA/IMS data. Pt-y of exposure were 290,355 for ETN, 502,646 Pt-y for INF, 531,959 Pt-y for LEF, and 15,675,396 Pt-y for MTX. Cannon GW, et al. Presented at: Annual Meeting of the ACR; October 16–21, 2004; San Antonio, Tex. Presentation 1469. Summary • AEs continue to be a concern for pts and clinicians • Hepatotoxicity/ALF is a concern, especially with MTX, but careful monitoring can prevent this from becoming a problem • TB rates in RA pts on TNF-α blockers seem to be higher than normal, but screening can decrease risk • Other opportunistic infections still seem to be of concern, so strong suspicion may prevent serious issues arising (cont) Drug Therapy • Paradigm shift – Inversion of conservative standard of care – DMARTs clearly improve patient outcomes by improving signs and symptoms decreasing pain and inflammation – DMARTs have been shown to retard x-ray progression – Thus the standard of care is to start aggressive therapy as soon as a certain diagnosis of progressive disease has been made – Includes combination chemotherapy as the best ongoing approach Drug Therapy • Even so – There is still no cure, real remissions are very rare – Ideally would prefer robust ACR 50 and 70 responses: not yet seen with monotherapy – The data from clinical trials really only approximate what may happen in the “real world”: is a 1 or 2 year data set reasonable to predict long term results over 20-30 years – Most patients need access to many possible therapies since there is no way to predict who might respond to any one therapy: thus it is important to have available as many potential therapies as possible with an acceptable benefit/risk ratio Treatment of RA • • • • • Clearly aggressive therapy with combinations of drugs is becoming standard of care Safety of these approaches is a significant issue The DMARTs are all good therapies with similar efficacy in monotherapy and in some patients even in combination There are still patients who are not adequately treated There is a significant safety signal with DMARTs including risk for infection, liver toxicity, ILD, lymphoma etc which require careful monitoring Summary • Many choices when selecting DMARDs (newly diagnosed and treated patients)—considerable body of data on which therapy/combination is most appropriate • However, close monitoring and early intervention, particularly with combination therapy, generally makes more clinical difference than the specific therapy chosen • While ACR20, and more recently ACR50 and 70 are good indicators of efficacy, other methods including SF36, HAQ and XR data are gaining in popularity as important tools for judging efficacy and measuring patient outcomes The Future • Limited by our successes – We have begun to believe our own press! Difficulty in recruiting patients who have done reasonably well on therapy; do we want to relegate studying new therapies only in those patients that fail the presently available therapies, are their results going to be broadly applicable to the general population with RA? – Many patients thus who will be available for study in the US will be those on the margin of society: those who cannot pay for drugs or are uninsured – Many studies will have to be done in Eastern Europe or elsewhere, given our inability to better define RA are we dealing with the same disease? – Why are there some pts who derive benefits regarding structural improvement but have few improvements in signs and symptoms? Trial Designs • Superiority trials against placebo, lower dose of test drug, active comparator • Equivalence trials, actually non –inferiority trials • effective comparative agents desired • more patients generally needed • Define margins of non inferiority • Other types of trials • induction Efficacy Outcomes • Efficacy must be “clinically meaningful” – Defined a priori – Raises question as to what is meant • Change in Sharpe score of two points • Clinicians like to talk about ACR 50 and 70 but regulatory approval threshold is ACR 20, but can you market a drug without a 50, 70? • Remission? Trial Designs • Placebo controlled with active comparator – Probably unethical for longer than 4 weeks – Last real development program to use: leflunomide • Classic “add-on” design – Used for cyclosporin, infliximab, adalimumab, abatacept, part of etanercept development program • Full factorial – Will define full benefit of study drug as well as safety also will reveal potential for synergy • Withdrawal design – Popular with pediatric trials – Variant of step down trials RA trials (safety) • Safety considerations may be different for drugs, biologics and devices • Cumulative incidences tend to be better • In new world of FDA, longer term exposures prior to approvals for safety • Use of comparator trials rather than observational experiences • Need to understand informed consent • Patient numbers; proposed 300 patients at highest dose for 6 months, 100 patients at highest dose for 1 year Some Possibilities • Approval Trials could be performed as non inferiority trials against presently available therapy since it is hard to imagine we will find drugs which are profoundly superior to the effect of those DMARTs presently available: BUT will marketing appreciate that? • Withdrawal trials of one drug vs another could be done as powerful tests of benefit, but companies and investors appreciate simpler superiority trials: they make better copy. Unmet Needs in RA • Reliable predictors of response to guide therapeutic decision making • Firm definition of therapy goals and clinically useful response markers • Effective therapy for current biologic failures or inadequate responders • Durable response (remission/cure) Unmet Needs (cont’d) • Reversal of pre-existing damage or earlier intervention to prevent damage • More affordable therapies • Best combination of agents is unknown • Safety concerns remain • Immunopathogenesis is not completely understood Donald G. Payan, M.D. Rigel’s Chief Scientific Officer and EVP Dr. Payan is one of Rigel’s co-founders and a member of the Board of Directors Dr. Payan has held senior level positions at numerous biotechnology companies, including AxyS Pharmaceuticals, Inc. and Khepri Pharmaceuticals, Inc He is also an Adjunct Professor of Medicine and Surgery at the University of California, San Francisco, is a former investigator in Howard Hughes Medical Institute, and is Board Certified in Allergy-Immunology and Infectious Diseases Rigel Product Pipeline Update: December 2005 R112: Allergic Rhinitis R343: Asthma (with Pfizer) R406/788: Rheumatoid Arthritis (and other immune diseases) R763: Oncology (with Serono) R017 Series: Transplant Rejection (and other immune/inflammatory diseases) Future Product Candidates Dr. Donald G. Payan 101 Rigel Product Pipeline Review December 2005 Build a strong and broad technology platform directed at the biology of disease mechanisms Focus on chronic illnesses that reflect significant diseases with large markets Leverage knowledge and expertise for each program across multiple clinical opportunities 102 Rigel R&D Goals Each year going forward: A First in Human (FIH) clinical study initiated / IND filing with a Rigel small molecule drug for a new target in an important disease 2004: R406/788 oral for Rheumatoid Arthritis (and other autoimmune diseases) 2005: R763 oral/IV in Oncology Going forward… 103 Rigel R&D Goals - Going Forward We have established the systems and organization to deliver one new molecule each year for an important FIH study initiated / IND filing 2006 Additional indications for R406/788 (ITP/AML/MS/SLE) R017 series for chronic transplant rejection 2007/08 R343 for Asthma w/Pfizer Rxxx for Axl Kinase in oncology/endometriosis 104 Rigel Product Pipeline December 2005 CLINICAL STAGE PHASE I PHASE II R112 inhaled Syk kinase inhibitor for Allergic Rhinitis R406/788 oral Syk kinase inhibitor for Rheumatoid Arthritis R763 oral/IV Aurora kinase inhibitor for Oncology PRE-CLINICAL IND-ENABLING PROGRAMS 2006 IND YEAR PROGRAM 2007 R017 series Oral JAK3 Inhibitor for Transplant Rejection R343 Inhaled Syk Kinase Inhibitor for Asthma (with Pfizer) 2nd Indication for R406/788 (ITP/AML/MS/SLE) Oral Axl Kinase Inhibitor for Oncology and Endometriosis DISCOVERY (ongoing lead molecule optimization) Virology - Hepatitis C (NS2 cysteine protease inhibitor; IRES inhibitor) Immunology/Inflammation - (TRAF-6 ligase inhibitor; PAK2 kinase inhibitor) Oncology - (PRK-1 kinase inhibitor) 105 Metabolism - (Adiponectin mimetics) PHASE III Rigel Pipeline Connections R406/788 R112 / R343 Airway Mucosal inflammation / atopic diseases Same target different cellular activities Chronic Tissue inflammation / destruction Allergic Rhinitis/Asthma Rheumatoid Arthritis/MS/SLE/ITP • 1º mucosal mast cells • Fcε - Syk kinase signaling • Multiple inflammatory mediators released • B Cells/macrophages/T cells • Fcγ - Syk kinase signaling • Multiple inflammatory mediators Same cells different cytokine target R017 series Chronic tissue rejection Transplant rejection • Bcells/Tcells - MLR • Jak3 kinase/cytokine signals • Specific immune cell proliferation 106 R763 Different growth factors and cellular responses Tumor Growth / progression Leukemia/Lymphoma/Solid tumor growth • Inhibit Transit through: • Specific phase of cell cycle • Selective growth factor responses • Aurora kinase inhibitor Allergic Rhinitis and Asthma: Common Mechanisms of Disease 107 IgE Production, Mast Cell Activation by Allergens, and Action of Mast Cell-Derived Mediators Singulair Lipid mediators (Leukotrienes, PGD2) Allergens Pollen Parasite Antigen presenting cell (Dendritic cell) Xolair CD4+ T cell B cell Plasma cell Acute allergic reaction (vasodilation, bronchoconstriction, mucus secretion) Antihistamines Th2 cell IL-4 (IL-13) Preformed mediators (Histamine, Proteases) Specific IgE production Basophil/Mast cell Late phase reaction (Lymphocyte, eosinophil, basophil, macrophage recruitment and activation) Chronic allergic inflammation (Structural and functional changes in tissue) 108 R112 R343 Allergen-induced receptor crosslinking Cytokines and chemokines (IL-4, TNF, IL-6, et al) Rigel’s Topical Syk Inhibitor in Allergic Rhinitis Primary Human Mast Cell Culture System Cell-based HTS IgE Signaling (200K cmpds) Med Chem MOA R112 Phase Ib • R112 appears effective Multiple Dose Safety Phase II Park Study • R112 is safe • R112 is fast and effective Allergic Rhinitis 109 Phase II 7-day comparative study Conclusions: R112 Park Study R112 is a selective inhibitor of Syk kinase - Discovered in a primary human mast cell HTS screen Blocks allergen triggered mast cell degranulation, leukotriene and prostaglandin secretion, and cytokine synthesis and release in the airway Desirable chemical and pharmaceutical characteristics - Ease of synthesis Shown to be safe with no adverse events The Phase II Park Study demonstrated efficacy (24% relative to placebo, p=0.0005) against all nasopharyngeal symptoms of allergic rhinitis when administered every 4 hours over 8 hours for 2 days as a nasal spray Rapid onset by 30 min with duration lasting throughout the study Complete study reported in JACI - April 2005 issue 110 Questions Raised by R112 Park Study How long does the beneficial effect of R112 last? Will it work in a less “disciplined” environment? (Fewer measurements; no supervision) How does it compare to IN Steroids? 111 Comparative Phase IIb R112 Trial in Allergic Rhinitis Initiated in August 2005 Randomized, double-blind study comparing R112 to placebo and Beconase AQ® nasal spray Primary Endpoints: • Efficacy as measured by total nasal symptom severity (TNSS) rating • Safety • Direct Steroid comparison in Allergy • Locations: 25 sites across the US • Patients: 396 verified to suffer from allergic rhinitis • Protocol: 7 days, twice a day treatment with R112, placebo or Beconase 112 Change from baseline in TNSS AM measurements Comparative Phase IIb R112 Trial in Allergic Rhinitis: Effects of R112 and Beconase on Total nasal symptom score (*TNSS) 0 -1 -2 -3 -4 -5 -6 -7 -8 0 1 2 3 4 5 6 7 Days of Treatment Twice/day: 113 R112 (n=139) Placebo (n=131) Beconase (n=63) *TNSS= sum of symptoms of congestion/runny nose/sneezing/itchy nose/post-nasal drip 8 Conclusions from Comparative Phase IIb R112 Trial in Allergic Rhinitis R112 did not significantly inhibit TNSS in allergy patients over a 7 day treatment period The positive control therapy with Beconase performed as expected An R112 follow-on in Allergic Rhinitis will need a longer duration of action (greater potency, slower dissolution rate) 114 Next Steps in Rigel Allergic Rhinitis Program Detailed evaluation of R112 clinical data and comparison of both Phase II studies Discussion with Pfizer regarding AR option (per contract) Potential selection of follow-on molecule with 100X> potency and slower dissolution rate (longer residency time in nose) 115 Rigel Pipeline Connections R406/788 R112 / R343 Airway Mucosal inflammation / atopic diseases Same target different cellular activities Chronic Tissue inflammation / destruction Allergic Rhinitis/Asthma Rheumatoid Arthritis/MS/SLE/ITP • 1º mucosal mast cells • Fcε - Syk kinase signaling • Multiple inflammatory mediators released • B Cells/macrophages/T cells • Fcγ - Syk kinase signaling • Multiple inflammatory mediators Same cells different cytokine target R017 series Chronic tissue rejection Transplant rejection • Bcells/Tcells - MLR • Jak3 kinase/cytokine signals • Specific immune cell proliferation 116 R763 Different growth factors and cellular responses Tumor Growth / progression Leukemia/Lymphoma/Solid tumor growth • Inhibit Transit through: • Specific phase of cell cycle • Selective growth factor responses • Aurora kinase inhibitor Syk Inhibitors: Potential Modulation of Both IgE or IgG Fc Receptor Signaling Anti-IgE Anti-IgG IgG IgE FcγR FcεR β Lyn P Syk Mast Cell Activation 117 FcRγ FcRγ Lyn P Syk Macrophage, B-lympocyte and Mast Cell Activation Rheumatoid Arthritis: A Progressive Debilitating Disease with Poor Current Treatment Options Swelling and inflammation 118 Rheumatoid Arthritis: A Progressive Debilitating Disease with Poor Current Treatment Options Swelling and inflammation Destruction of cartilage 119 Rheumatoid Arthritis: A Progressive Debilitating Disease with Poor Current Treatment Options Swelling and inflammation Destruction of cartilage Bone deterioration DMARDs (Disease-Modifying Anti-Rheumatic Drugs) introduced 120 Rheumatoid Arthritis: A Progressive Debilitating Disease with Poor Current Treatment Options Swelling and inflammation Destruction of cartilage Bone deterioration DMARDs (Disease-Modifying Anti-Rheumatic Drugs) introduced Destruction slows 121 R406/788 for Rheumatoid Arthritis R406/788 122 Designed to prevent bone and cartilage damage R406/788 Inhibits Several Steps that Initiate and Maintain RA Disease Activity Periphery Joint Synovium Bone/Cartilage R406 I 3 ,1 0 1 4, L B Immune Complex BCR R406 CD40 CD40L T FcγR R406 Mast MMP Synovial Fibroblast TCR PGE2 H+ CatK MHC FcγR FcγR TNF IL-1 R406 Osteoclast FcγR IL-6 Macrophage O2- IL-17 IL-8 FcγR PMN KC VEGF Pannus Disease Initiation Disease Propagation Tissue Damage R406 Has Demonstrated Efficacy in Three Different Animal Models of Rheumatoid Arthritis 1) Mouse Collagen Antibody Induced Arthritis (CAIA) 2) Rat Collagen Induced Arthritis (CIA) 3) Mouse K/B x N Spontaneous Autoantibody Induced Arthritis All three models display tissue swelling and bone destruction in and near affected joints resulting in loss of mobility and function R406 reversed all these changes in all three models 124 R406 Pharmacology: Efficacy in the Rat Collagen Induced Arthritis (CIA) Model Radiographs 8 Vehicle Control - Day 28 Hind Paw clinical score a c i n i l C w a P d n i H 7 6 5 Vehicle 10 mg/kg b.i.d 30 mg/kg b.i.d 4 3 R406 - Day 28 2 1 0 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 Days Post Arthritis Onset Immunization Day Day-post onset 125 Collagen type II+IFA i.d. CII/IFA boost 0 7 Treatment with R406 b.i.d. from onset (clin. score>1) * 10-18 0 Clinical score/histology, Hind-paw thickness, Body weight, Cytokine/chemokine Radiographic score 18 R406 Prevents Joint and Bone Destruction in Rat Collagen Arthritis Model Rheumatoid Arthritis No Treatment (Vehicle) 126 Data from E. Brahn, MD, UCLA Rheumatoid Arthritis Treated with R406 30 mg/kg R406 Phase I Clinical Study Overview (in normal volunteers) Title: • A Single Center, Phase I, Double-Blind, Randomized, PlaceboControlled Study to Investigate the Safety, Tolerance and Pharmacokinetic Profile of Single and Multiple Doses of R406 in Young Healthy Male Subjects Design: • Single Dose: 5 sequentially rising doses (80, 250, 400, 500, and 600 mg) 5-6 R406 and 1-2 placebo subjects per group • Multiple Dose: 3 sequentially rising doses (100, 200, and 300 mg bid) given for 7 days 6 R406 and 2 placebo subjects per group 127 R406 Phase I Clinical Study Details Safety • Adverse event profile • Vital signs • ECG (lead II & 12-lead) • Clinical laboratory safety tests (chemistry, hematology, urinalysis) Pharmacokinetics • In multiple dose phase, subjects were administered a single dose on Days 1 and 7 and two doses per day on Days 2-6, to allow for optimal PK profiling Pharmacodynamics • • • 128 Activated Basophil Assay (FACS, CD63+ve), Fcε signaling FACS analysis of peripheral blood Platelet aggregation R406 Phase I Results – Safety Single Dose Phase • All doses levels well tolerated, with the highest dose of 600 mg associated with an increased frequency of postural dizziness • No other significant findings, including vital signs, ECG, laboratory testing (hematology,chemistry, urinalysis) Multiple Dose Phase • 100 and 200 mg bid Well tolerated; no significant findings including vital signs, ECG, laboratory testing • 300 mg bid Several adverse events of moderate severity, including increased standing diastolic BP, a variety of mild GI-related symptoms, and headache Hematology No 129 findings included ↓ Neutrophils significant findings in chemistry, urinalysis, or ECG testing Activated Basophil Assay: Biomarker for R406 activity in patient’s blood Demonstrate effects on the Fcε pathway, based on the commercially available Basotest assay Human peripheral blood basophils stimulated with anti-IgE ex vivo undergo degranulation and express CD63 (gp53, LAMP-3) on the cell surface. CD63 is readily detected by anti-CD63-FITC specific antibodies using flow cytometry. Basophils are identified by positive staining with a PE labeled anti-IgE antibody Reference range is 15-60% activated (CD63+) basophils, however, some subjects were low responders 130 Basophil Activation Assay demonstrates activity of R406 in blood of normal volunteers Single Dose Group Means % Activated Basophils 60 Placebo 40 80 mg 250 mg 400 mg 20 500 mg 600 mg 0 0 4 8 12 16 Time Post-Dose (h) 131 20 24 PK/PD Correlation, Basophil Activation Assay for R406 in normal volunteers 160 140 120 Observed PD (%) 100 Predicted 80 60 40 20 0 1 10 100 1000 10000 Plasma Conc.of R940406 (ng/mL) Parameter Emax EC50 Unit Estimate Standard Error CV (%) 84.2 3.82 4.53 496 42.2 8.51 2.25 0.19 8.54 % Control * ng/mL Gamma * Parameters were obtain using a Sigmoidal 132 -Emax model, with 1/y weighting. Average plasma concentration for R406 in normal human volunteers when R406 was orally administrated 8000 R940406 plasma conc. (ng/mL) 7000 6000 5000 100 mg Day 7 200 mg Day 7 300 mg Day 7 4000 3000 2000 1000 0 144 154 164 174 184 Time (h) 133 Time shown in # of hours 194 204 214 R406 Phase I Results – Human PK Comparison with Rat CIA R406 Plasma concentration (ng/mL) 10000 200 mg (2.5 mg/kg) Human (Day 7) 10 mg/kg, Rat CIA model (Day 18) 30 mg/kg, Rat CIA model (Day 18) 1000 100 Efficacy in the Rat CIA 10 1 0 2 4 6 8 10 12 14 Time Post-Dose (h) Time (hours) 16 18 Hind Paw score Hindclinical Paw Clinical Score 8 Vehicle 7 10 mg/kg b.i.d 6 30 mg/kg b.i.d 5 4 3 202 22 24 1 0 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 Days Post Arthritis Onse t 134 Conclusions of R406/788 as an Oral RA Treatment R406/788 is a potent and selective inhibitor of Syk R406/788 blocks FcγR signaling on mast cells and macrophages, and BCR signaling R406/788 is orally bioavailable in all species tested Efficacy demonstrated in multiple autoimmune disease models reduction in multiple inflammation cytokines and tissue protection Good safety profile in animal models Clinical studies underway - promising pharmacodynamics in Phase I May be useful for several disease indications (ITP, AML, MS, SLE) 135 Next Steps in RA for R406/788 Evaluate safety and PK in single and multiple dose / drug-drug interaction studies (eg. Methotrexate) Evaluate preliminary efficacy and safety in patients with rheumatoid arthritis - 2H 2006 Identify additional clinical indications for R788 in autoimmune diseases Rigel to continue program through NDA 136 R406/788 in Several Disease Areas: leverage molecule across multiple clinical opportunities Target cells MOA Inhibition by R406 (EC50) Allergy/Asthma Mast cells B cells Disease area Ig-FcγR/Syk 30-60 nM Macrophages Autoimmune diseases (MS/SLE/ITP) Inflammatory diseases Leukemic cells Dendritic cells 137 Leukemia FLT3L/FLT3R 10-80 nM Autoimmune diseases (MS) R788 in ITP Immune Thrombocytopenia Purpura (ITP): autoimmune hematological disease characterized by destruction of platelets Prevalence of approx. 200,000 in US More women than men; half new cases in children Current therapy inadequate -- steroids mainstay • Platelet destruction mediated by IgG signaling • R788 potent inhibitor of IgG signaling • R788 shown to protect from thrombocytopenia in animal models 138 R788 in ITP R788… Improves thrombocytopenia in ITP mouse model Does not affect murine bleeding time Does not affect platelet aggregation in humans in response to collagen Phase I studies nearly completed 139 R788 in Acute Mylelogenous Leukemia (AML) Flt3-mutant AML • Approximately 20-30%of AML is Flt3-mutant • Flt3-mutant AML patients have poor prognosis R788 also potently inhibits Flt3 R788 inhibits tumor growth of human Flt3 AML cells in mouse xenografts R788 extends survival in mouse model of leukemia 140 R788 in Flt3 AML Survival Percent survival 100 Vehicle 20mg/kg 40mg/kg 80mg/kg 75 >51% ILS 50 45% ILS P<0.01 Dosing began day 14 post-implantation 25 Tumor Shrinkage 15 13% ILS Vehicle 20mg/kg R788 bid 40mg/kg R788 bid 0 25 50 Day 75 100 Change inTumor Volume (V/V 0) 0 10 %T/C = 35.1 5 0 %T/C = -0.4 0 5 10 15 Day 141 20 25 30 R788: Next Steps in ITP/Flt3 AML/Autoimmune Diseases Complete Phase I safety studies 1H 2006 Initiate clinical trials in 2H 2006 Obtain proof of concept clinical data late 2006 Animal model studies underway in other more chronic autoimmune diseases (MS/SLE) 142 Rigel Pipeline Connections R406/788 R112 / R343 Airway Mucosal inflammation / atopic diseases Same target different cellular activities Chronic Tissue inflammation / destruction Allergic Rhinitis/Asthma Rheumatoid Arthritis/MS/SLE/ITP • 1º mucosal mast cells • Fcε - Syk kinase signaling • Multiple inflammatory mediators released • B Cells/macrophages/T cells • Fcγ - Syk kinase signaling • Multiple inflammatory mediators Same cells different cytokine target R017 series Chronic tissue rejection Transplant rejection • Bcells/Tcells - MLR • Jak3 kinase/cytokine signals • Specific immune cell proliferation 143 R763 Different growth factors and cellular responses Tumor Growth / progression Leukemia/Lymphoma/Solid tumor growth • Inhibit Transit through: • Specific phase of cell cycle • Selective growth factor responses • Aurora kinase inhibitor Organ Transplantation Overview Approximately 90,000 organ transplants worldwide 27,022 transplants in the U.S. in 2004 with 6% growth First year survival rates are ~90% for the major organs Chronic rejection rates are high (~50% at 5-10 years) Conventional maintenance therapy consist of 3 drug combo: >Calcineurin inhibitor (NeoralTM or PrografTM) >Mycophenolate mofetil (CellceptTM) >Corticosteroids 144 Transplantation Pharma Market Current Global pharma sales approx. $4B TM > Cellcept (Roche)= $1.1B TM > Prograf (Fujisawa)= $1.1B TM > Neoral (Novartis)= $1.0B Current drugs exhibit non-immune specific toxicities Current Focus > Reduce calcineurins (Neoral/Prograf) and steroid use > Better balance between toxicity and immune suppression Current Patient Needs > Reduce toxicities (1º nephrotox, 2° hypertension, neurotox, hyperlipidemia) > Prevention of chronic rejection and vasculopathy 145 Pathogenesis and Treatments of Transplant Rejection NK Cell CD8+ T cells 5 IL15 5 IL7 Allograft Progenitor 2 B7.1/B7.2 CD28 IL2 5 3 MHC DC IL2 5 T Cell Proliferation 4 IL4 TCR 1 5 T CD40L B 1 OKT3 B B 2 CTLA4-Ig 3 CsA/FK506 4 MMF, azathioprine 5 Cytokine signaling inhibitors - Rapamycin 146 Alloantibody Production The IL-2 and IL-4 Pathway: Key Signals in immune mediated rejection IL-2 IL-4 γc IL-2Rα P P Y phosphorylation P S/T phosphorylation JAK1 JAK3 γc IL-4Rα P JAK1 JAK3 IL-2Rβ P IRS P P STAT3/5 Shc P STAT6 Grb2 Accessory Pathways Sos PI3K STAT3/5 P P ERK Akt STAT3/5 P Modulatory Pathways P38, Erk, other kinases? P Nuclear Translocation STAT3/5 P P STAT3/5 P P Gene Transcription Janus Tyrosine Kinase 3 A cytoplasmic tyrosine kinase Expression limited to T & B cells, natural killer (NK) cells, mast cells, and macrophages Activated by multiple cytokines, including IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21 Plays a critical role in lymphocyte development and function Attractive target based on limited tissue distribution 148 Properties of Rigel Jak3 inhibitor - R017 series Potent: <100nM in cell-based assays of T-cell, IL4, IL2 signaling and MLR Selective against IFNγ and Epo signaling Good selectivity against other off target assays HERG ≥ 12µM Ames negative Soluble: > 20mg/ml in water Good exposure and sustained levels in animal models Good distribution to target tissues Activity in the mouse DTH model Well tolerated up to 80mg/kg/day in Lewis rat 149 Heterotopic Rat Heart Transplant Allograft Model Brown Norway (BN) male hearts are grafted onto the abdominal region of Lewis (LW) male rats Vascularized Model Graft Heart Ischemia to Reperfusion about 30 min BN to LEW rats MHC RI is mismatched, robust immune response Average Rejection Day 7+/- 1 without treatment Pfizer compound CP-690,550 tested positive in murine model (DBA2 to C57/Bl6)75% survival at D28 (140 ng/ml blood level) Most commonly used animal model in transplantation Rapamycin, CsA, FK506 can completely sustain graft survival in rat allograft model 150 R017 prolongs Heterotopic Heart Graft Survival 100 % Grafts Survival 80 60 Vehicle (Allograft) (n=3) (BN Vehicle (Isograft) (n=6) (LW LW) R017 (Allograft) (20 mg/kg) b.i.d. (n=6) (BN R017 (Allograft) (40 mg/kg) b.i.d (n=6) (BN 40 20 0 151 LW) 0 4 8 12 16 Time(Days) 20 24 28 LW) LW) Global effect of R017 on Grafted Hearts 152 LW LW BN LW vehicle/untreated BN LW 40mg/kg R017 LW LW BN LW vehicle/untreated BN LW 40mg/kg R017 Conclusions from Rat Heart Transplantation Study with R017 Rats treated with R017 at 40 mg/kg maintain robust heart graft function, prolongs graft survival for at least 28 days (6/6) 20 mg/kg bid of R017 prolongs allograft function (5/6) for at least 28 days Dose Response of Efficacy with R017 at 40 mg/kg and 20 mg/kg: Gross appearance of grafts, heart graft contraction by palpation, and ECG recordings 153 R017 series Jak3 inhibitor in Transplantation: Status and 2006 plans Lead molecule, R017, validated in animal model of cardiac transplantation Select clinical candidate, from R017 series, in 1H 2006 Complete IND enabling studies in 2H 2006 Initiate FIH studies / file IND in 1H 2007 154 Rigel Pipeline Connections R406/788 R112 / R343 Airway Mucosal inflammation / atopic diseases Same target different cellular activities Chronic Tissue inflammation / destruction Allergic Rhinitis/Asthma Rheumatoid Arthritis/MS/SLE/ITP • 1º mucosal mast cells • Fcε - Syk kinase signaling • Multiple inflammatory mediators released • B Cells/macrophages/T cells • Fcγ - Syk kinase signaling • Multiple inflammatory mediators Same cells different cytokine target R017 series Chronic tissue rejection Transplant rejection • Bcells/Tcells - MLR • Jak3 kinase/cytokine signals • Specific immune cell proliferation 155 R763 Different growth factors and cellular responses Tumor Growth / progression Leukemia/Lymphoma/Solid tumor growth • Inhibit Transit through: • Specific phase of cell cycle • Selective growth factor responses • Aurora kinase inhibitor Aurora A and B are Essential for Proper Chromosome Segregation During Mitosis (cell division) Prophase Anaphase Telophase Metaphase A - centrosomes A - spindle pole microtubules B - centromere A - polar microtubules B - spindle midzone, cell cortex at site of cleavage furrow Aurora A Aurora B Centrosome separation and maturation Spindle assembly Kinetochore-microtubule interactions Spindle assembly checkpoint Cytokinesis Carmena and Earnshaw, Nat Rev Mol Cell Biol 2003. 4, 842-854. 156 A - midbody B - midbody Inhibition of Aurora A and B Causes Cell Death And/or Endoreduplication Apoptotic nuclei Cell death No cell division Endoreduplication Mitotic cell Aurora Kinase Inhibitors Absence of cytokinesis G2 S 157 M G1 Enlarged Nuclei Biological Properties of Rigel’s Aurora Kinase Inhibitor R763 Potent antiproliferative effect across a broad panel of cell types Good selectivity over cell-based counter assays Phenotype consistent with inhibition of Aurora kinases: - Endoreduplication, i.e., exit from mitosis without cytokinesis and cell cycle arrest Induces apoptosis No effect on quiescent primary hepatocytes Aurora B expression may correlate with R763 sensitivity in some tumors Inhibition of blood vessel formation (angiogenesis) 70 gene “signature” identified as markers to discriminate R763 sensitive and resistant Tumors: more focused patient selection possible 158 R763 in Leukemia Models Effects of R763 on Tumor Growth in Leukemia cell line (MV411) Mouse Xenograft Model Schedule Monitor Tumor growth, body weight, and clinical observations 24 days Day 0 Day 28 Day 70+ Animals remaining 12.5 mg/kg n=1 20 mg/kg n=5 Cell injection into NOD SCID mice (Subcutaneous) Start treatment Mean tumor volume=333mm3 160 Stop treatment Total treatment duration = 4 cycles of 3 on/4 off Effects of R763 on Tumor Growth in Leukemia cell line (MV411) Mouse Xenograft Model 2500 1500 Percent Survival 2000 Tumor Volume (mm3) 100 R763 Vehicle Cytarabine Vehicle Cytarabine, 100mg/kg R763, 7.5 mg/kg R763, 10 mg/kg R763, 12.5 mg/kg R763, 20 mg/kg 1000 75 50 25 500 0 0 0 2 Treatment Phase 161 4 6 8 10 12 14 16 18 20 22 24 26 28 0 10 20 30 40 50 Study Day Treatment Phase Days on Study 30 R763 Dose Level %ILS Tumor Growth % partial %complete Apparent net tumor Delay (days) regression regression cell kill (logs) 7.5 mg/kg 100 19.7 0 0 -0.4 10 mg/kg 143 27.4 67 0 0 12.5 mg/kg 172 32.3 92 0 0.3 20 mg/kg 223 42.8 83 17 0.9 60 70 Status of Rigel Aurora Kinase & R763 program From HTS hit, to filing of the R763 IND: 28 months Partnership with Serono established October 2005, to develop R763 for the treatment of cancer Rigel will potentially receive up to $160 million in payments, including: • Upfront payment of $25 million in cash and equity • Milestone payments of $135 million • Royalties on sales Rigel to file an IND for R763 - December 2005 Serono will initiate clinical trials in 2006 Serono will develop and commercialize R763 and follow-up compounds 162 Rigel Pipeline Connections R406/788 R112 / R343 Airway Mucosal inflammation / atopic diseases Same target different cellular activities Chronic Tissue inflammation / destruction Allergic Rhinitis/Asthma Rheumatoid Arthritis/MS/SLE/ITP • 1º mucosal mast cells • Fcε - Syk kinase signaling • Multiple inflammatory mediators released • B Cells/macrophages/T cells • Fcγ - Syk kinase signaling • Multiple inflammatory mediators Same cells different cytokine target R017 series Chronic tissue rejection Transplant rejection • Bcells/Tcells - MLR • Jak3 kinase/cytokine signals • Specific immune cell proliferation 163 R763 Different growth factors and cellular responses Tumor Growth / progression Leukemia/Lymphoma/Solid tumor growth • Inhibit Transit through: • Specific phase of cell cycle • Selective growth factor responses • Aurora kinase inhibitor Rigel Product Pipeline December 2005 CLINICAL STAGE PHASE I PHASE II R112 inhaled Syk kinase inhibitor for Allergic Rhinitis R406/788 oral Syk kinase inhibitor for Rheumatoid Arthritis R763 oral/IV Aurora kinase inhibitor for Oncology PRE-CLINICAL IND-ENABLING PROGRAMS 2006 IND YEAR PROGRAM 2007 R017 series Oral JAK3 Inhibitor for Transplant Rejection R343 Inhaled Syk Kinase Inhibitor for Asthma (with Pfizer) 2nd Indication for R406/788 (MS/SLE/ITP/AML) Oral Axl Kinase Inhibitor for Oncology and Endometriosis DISCOVERY (ongoing lead molecule optimization) Virology - Hepatitis C (NS2 cysteine protease inhibitor; IRES inhibitor) Immunology/Inflammation - (TRAF-6 ligase inhibitor; PAK2 kinase inhibitor) Oncology - (PRK-1 kinase inhibitor) 164 Metabolism - (Adiponectin mimetics) PHASE III Validation of Rigel Drug Discovery Approach Through Large Pharma Partnerships Asthma/COPD (2005) Immune Diseases (1999) Oncology (1998) Angiogenesis (2001) Immune Diseases (1999) Ligase Target in Oncology (2002) Ligase Targets in Oncology (2004) Aurora Kinase Inhibitors in Oncology (2005) 165 Questions and Answers Rigel NASDAQ:RIGL