Enhancing the Lives of Patients with Heme Malignancies - Corporate Presentation March 2023
←
→
Page content transcription
If your browser does not render page correctly, please read the page content below
Forward-Looking Statements and Safe Harbor Except for the historical information contained herein, this presentation contains forward-looking statements made pursuant to the “safe harbor” provisions of the Private Securities Litigation Reform Act of 1995. Investors are cautioned that such statements, include, without limitation, those regarding: (i) that Geron plans to submit regulatory filings in the U.S. in mid-2023 and the EU in the second half of 2023, and is preparing for a potential regulatory approval and commercial launch in lower risk MDS in the U.S. in the first half of 2024 and in the EU by the end of 2024; (ii) that for IMpactMF, Geron expects to conduct an interim analysis in 2024 and a final analysis in 2025; (iii) that for the next generation telomerase inhibitor program, the Company expects completion of the current discovery effort in 2023 and potentially advance any lead compounds into the next step of discovery research; (iv) that Geron expects its projected financial resources to fund operations, including completion of preparatory activities for potential U.S. commercial launch of imetelstat in lower risk MDS, until the third quarter of 2025; (v) that IMerge Phase 3 and IMpactMF have registrational intent; (vi) that there is clinical and molecular evidence supporting the potential for MDS disease modification with imetelstat and that imetelstat also has the potential to demonstrate disease- modifying activity in patients by reducing the malignant stem and progenitor cells of the underlying disease; (vii) that the Company expects imetelstat to be a highly differentiated product in the lower risk MDS commercial marketplace; (viii) that the Company projects that the addressable patients in 2030 for imetelstat in LR MDS are approximately 33,000 and for R/R MF are approximately 18,000; (ix) that the Company believes imetelstat has potential large market opportunities with potential peak 2030 market opportunity from the U.S. and key European markets of approximately ~$3 billion, with ~$1.2 billion from LR MDS sales and ~$1.8 billion from R/R MF sales; (x) that there are unmet needs in LR MDS and R/R MF potentially addressed with imetelstat treatment; (xi) that the telomerase inhibition of imetelstat gives it the potential for expanding into new indications; (xii) that the Company the planned first site opening for IMpress is in 2023; (xiii) that the Company expects that the TELOMERE Phase 1 clinical trial will be initiated pending data from the IMpress Phase 2 clinical trial; (xiv) that the Company expects further experiments from the preclinical program in lymphoid malignancies; and (xv) other statements that are not historical facts, constitute forward-looking statements. These forward-looking statements involve risks and uncertainties that can cause actual results to differ materially from those in such forward-looking statements. These risks and uncertainties, include, without limitation, risks and uncertainties related to: (a) the impact of general economic, industry or political climate in the U.S. or internationally, the current or evolving effects of macroeconomic conditions, such as the COVID-19 pandemic, civil or political unrest or military conflicts around the world, such as the military conflict between Ukraine and Russia, inflation, rising interest rates or prospects of a recession, on Geron’s business and business prospects, its financial condition and the future of imetelstat; (b) whether Geron overcomes all of the potential delays and other adverse impacts caused by the current or evolving effects of the COVID-19 pandemic and/or geopolitical events, as well as all the enrollment, clinical, safety, efficacy, technical, scientific, intellectual property, manufacturing and regulatory challenges in order to have the financial resources for, and to meet the expected timelines and planned milestones in (i) to (iii), (ix) and (xii) to (xiv) above; (c) whether regulatory authorities accept for filing Geron’s planned NDA and MAA submissions and permit the further development of imetelstat on a timely basis, or at all, without any clinical holds; (d) whether imetelstat is demonstrated to be safe and efficacious in IMerge Phase 3 and IMpactMF to enable regulatory approval; (e) whether any future efficacy or safety results may cause the benefit-risk profile of imetelstat to become unacceptable; (f) whether imetelstat actually demonstrates disease- modifying activity in patients and the ability to target the malignant stem and progenitor cells of the underlying disease; (g) that Geron may seek to raise substantial additional capital in order to complete the development and commercialization of imetelstat and to meet all of the expected timelines and planned milestones in (i) to (iii), (ix) and (xii) to (xiv) above; (h) whether regulatory authorities require additional clinical testing of imetelstat prior to or after granting approval in lower risk MDS even though IMerge Phase 3 met its primary endpoint; (i) whether there are failures in manufacturing or supplying sufficient quantities of imetelstat that would delay, or not permit, the anticipated launches in (i) above or not enable the other ongoing or planned clinical trials; (j) whether imetelstat is able to obtain and maintain the exclusivity terms and scopes provided by patent and patent term extensions, orphan drug, data and marketing and pediatric coverages and have freedom to operate; (k) whether the follow-up period of 12 months for the IMerge Phase 3 primary analysis results in not obtaining adequate data to demonstrate safety and efficacy, including transfusion independence, in the primary analysis; (l) whether Geron can accurately project the timing of complete enrollment in its clinical trials, whether due to the current or evolving effects of the COVID-19 pandemic and/or geopolitical events or otherwise; (m) whether Geron is able to enroll its clinical trials at a pace that would enable the financial resources for, and to meet the expected timelines and planned milestones in (i) to (iii), (ix) and (xii) to (xiv) above; (n) that Geron may be unable to successfully commercialize imetelstat due to competitive products, or otherwise; (o) if the FDA does not grant priority review to the IMerge data, then the launch date in lower risk MDS may be later than the first half of 2024; (p) whether Geron may decide to partner and not to commercialize independently in the U.S. and in key European markets; and (q) for IMpactMF, Geron’s projected rates for enrollment and death events may differ from actual rates, which may cause the interim analysis to occur later than 2024 and the final analysis to occur later than 2025. Additional information on the above risks and uncertainties and additional risks, uncertainties and factors that could cause actual results to differ materially from those in the forward-looking statements are contained under the heading “Risk Factors” or other similar headings found in documents Geron files from time to time with the Securities and Exchange Commission (the “SEC”), including the Company’s Report on Form 10-K for the year ended December 31, 2023 and subsequent filings. Undue reliance should not be placed on forward-looking statements, which speak only as of the date they are made, and the facts and assumptions underlying the forward-looking statements may change. Except as required by law, Geron disclaims any obligation to update these forward-looking statements to reflect future information, events or circumstances. 2
Geron Today Positive Phase 3 Top-Line Catalyst-Rich Multi-Billion Dollar Results in Lower Risk MDS Next Two Years Expected Market Potential Statistically significant and 2023 U.S. NDA and EU MAA Expected imetelstat profile to meet clinically meaningful submissions for LR MDS unmet needs in LR MDS and R/R MF improvements with imetelstat vs. placebo: Commercial preparations ongoing • Primary 8-week transfusion 2024 U.S. commercial launch • Hiring experienced senior leadership independence (TI) endpoint and key of imetelstat in LR MDS* secondary 24-week TI endpoint met • Executing “go-to-market” plan • Substantial increases in hemoglobin Interim analysis overall • Engaging stakeholders for marketplace levels and reductions in transfusions survival Phase 3 readout in readiness • Efficacy across key MDS subtypes, R/R MF including RS+ and RS- patients Potential market opportunity 2023-25 Significant life cycle • >$3B combined potential peak market Safety results consistent with prior opportunity for LR MDS and R/R MF management program imetelstat clinical experience readouts Clinical and molecular evidence supporting the potential for disease modification * If the NDA is accepted for filing and imetelstat is approved for commercialization by the FDA References on slide 52 3 LR MDS = lower risk myelodysplastic syndromes; NDA = new drug application; MAA = marketing authorization application; R/R MF = relapsed/refractory myelofibrosis; RS+ = ring sideroblast positive; RS- = ring sideroblast negative
Anticipated Momentum Going Forward 2024 IMpactMF Ph3 1H 2024 interim analysis for Jan 4, 2023 Mid-2023 LR MDS median overall survival (OS) in Positive IMerge LR MDS U.S. approval & relapsed/ U.S. NDA commercial launch* refractory MF Ph3 LR MDS TLR submission reported 2H 2023 2H 2H 2024 2024 LR MDS LR MDS EU MAA LR MDS EU approval & EU approval & submission commercial commercial launch* launch commercial launch* If the NDA / MAA are accepted for filing and imetelstat is approved for commercialization by the FDA / EMA, as applicable LR MDS = lower risk myelodysplastic syndromes; TLR = top-line results; NDA = new drug application; MAA = marketing authorization application; MF = myelofibrosis 4
Novel Target, Novel Drug Telomerase inhibition and imetelstat to potentially alter the course of hematologic malignancies 5
Telomerase, a Unique Oncology Target Malignant HSCs are the malignant Heme source of disease in heme hematopoietic malignancies stem cells malignancies • Malignant hematopoietic Myelodysplastic stem cells (HSCs) give rise Syndromes (MDS) to uncontrolled proliferation of malignant blood cells • Malignant HSCs rely on continual upregulation of Telomerase Myelofibrosis (MF) telomerase to support continually uncontrolled proliferation upregulated Telomerase is a novel Acute Myeloid Leukemia (AML) target in malignantly uncontrolled proliferation transformed HSCs of malignant cells (malignant hematopoiesis) References on slide 52 6
Imetelstat, First-in-Class Telomerase Inhibitor Imetelstat enables recovery of bone marrow Imetelstat binds to telomerase and and blood cell production inhibits its activity Evidence for potential disease modification: • Clinical efficacy results: durable TI (LR MDS); improved OS (R/R MF) • Molecular data: reductions in variant allele apoptosis of frequency (VAF) malignant cells • Depletion of mutated abnormal malignant cells that have been associated with the malignant disease, such as SF3B1, ASXL1, JAK2V617F and CALR TI = transfusion independence; LR MDS = lower risk myelodysplastic syndromes; OS = overall survival References on slide 52 7
Phase 3 IMerge trial design Phase 3 Double blind, randomized Imetelstat 118 clinical sites in 17 countries 7.5mg/kg every 4 weeks Primary Endpoint: (n=118) 8-week RBC Transfusion Independence (TI)** Patient Population ( ITT n=178): R Stratification: • Transfusion burden (4 - 6 vs. >6 units) Key Secondary • IPSS Low- or Intermediate 1- Risk MDS A • Relapsed/Refractory* to ESA or EPO N • IPSS risk category (Low vs. Intermediate-1) Endpoints: D Supportive care, including RBC and platelet • 24-week RBC TI** >500 mU/ml transfusions, myeloid growth factors (e.g., G-CSF), and O 2:1 • Duration of TI • Transfusion dependent: ≥4 units RBCs M iron chelation therapy administered as needed on study every 8 weeks over 16-week pre-study I per investigator discretion • Hematologic Improvement- • Non-deletion 5q Z Erythroid (HI-E) • No prior treatment with lenalidomide E Placebo or HMAs (n=60) * Received at least 8 weeks of ESA treatment (epoetin alfa ≥40,000 U, Safety population (treated) n=177 ** Proportion of patients without any RBC epoetin beta ≥30,000 U or darbepoetin alfa 150 mcg or equivalent per transfusion for at least eight consecutive weeks week) without Hgb rise ≥1.5 g/dL or decreased RBC transfusion Imetelstat n=118 since entry to the trial (8-week TI); proportion of requirement ≥ 4 units/8 weeks or transfusion dependence or reduction patients without any RBC transfusion for at least in Hgb by ≥1.5 g/dL after hematologic improvement from ≥ 8 weeks of Placebo n=59 24 consecutive weeks since entry to the trial (24- ESA treatment. week TI) EPO = erythropoietin; ESA = erythropoietin stimulating agents; G-CSF = granulocyte colony stimulating factor; Hgb = hemoglobin; IPSS = International Prognostic Scoring System; ITT = intent to treat; RBC = red blood cell; HI-E = hematologic improvement-erythroid; HMAs = hypomethylating agents; MDS = myelodysplastic syndromes 9 References on slide 52
Key trial metrics Balanced time on study and treatment time between two arms Imetelstat Placebo (n=118) (n=60) Median time on study, months (range) 19.5 (1.4-36.2) 17.5 (0.7-34.3) Median time on treatment, months (range) 7.8 (0.03-32.5) 6.5 (0.03-26.7) Median time on treatment for 8-week TI responders, months (range) 17.2 (1.8-32.5) 15.0 (4.1-25.8) Median treatment, cycles (range) 8 (1-34) 8 (1-30) Median treatment cycles for 8-week TI responders, months (range) 18 (3-34) 17 (3-29) References on slide 52 8-week TI = proportion of patients without any RBC transfusion for at least eight consecutive weeks since entry to the trial TI = transfusion independence 10
Baseline patient demographics and disease characteristics Comparable between both arms Imetelstat Placebo (n=118) (n=60) Age, years, median (range) 71.5 (44-87) 73.0 (39-85) WHO 2001 category, n (%) RS+ 73 (61.9) 37 (61.7) RS- 44 (37.3) 23 (38.3) RBC transfusion burden, units/8 weeks, median (range) 6 (4-33) 6 (4-13) 4 - 6 units / 8 weeks, n (%) 62 (52.5) 33 (55.0) >6 units / 8 weeks, n (%) 56 (47.5) 27 (45.0) IPSS risk category, n (%) Low 80 (67.8) 39 (65.0) Intermediate-1 38 (32.2) 21 (35.0) Prior luspatercept use, n (%)* 7 (5.9) 4 (6.7) Pre-treatment hemoglobin**, median (range), g/dL 7.9 (5.3-10.1) 7.8 (6.1-9.2) * Insufficient number of patients previously treated with luspatercept to draw conclusions about the effect of imetelstat treatment in such patients. None of the imetelstat-treated patients and one of the placebo patients achieved 8-week TI. ** Pretreatment hemoglobin is defined as the average of all hemoglobin values in the eight weeks prior to the first dose date, excluding values that were within 14 days after transfusion (thus considered to be influenced by transfusion). References on slide 52 11 RS+ = ring sideroblast positive; RS- = ring sideroblast negative; WHO = World Health Organization
Patient disposition after 18 months median follow up Imetelstat Placebo (n=118) (n=59) Treatment ongoing, n (%) 27 (22.9) 14 (23.7) Treatment discontinued, n (%) 91 (77.1) 45 (76.3) Lack of efficacy 28 (23.7) 25 (42.4) Adverse event 19 (16.1) 0 Cytopenias 11 (9.3) 0 Unrelated 8 (6.8) 0 Disease relapse after initial response on study 17 (14.4) 1 (1.7) Patient decision 16 (13.6) 10 (16.9) Progressive disease 7 (5.9) 5 (8.5) AML progression 2 (1.7) 1 (1.7) Investigator decision 2 (1.7) 2 (3.4) Death* 1 (0.8) 2 (3.4) Lost to follow up 1 (0.8) 0 * On imetelstat treatment arm, patient death: neutropenic sepsis not related to drug after ~ two-year treatment duration On placebo treatment arm, patient deaths: (1) COVID-19 and (1) heart valve issue References on slide 52 12 AML = acute myeloid leukemia
Top-Line Efficacy Results Primary and key secondary endpoint met with high statistical significance and clinically meaningful improvements 13
Met primary endpoint (8-week TI) Highly statistically significant and clinically meaningful improvement in 8-week TI Imetelstat Placebo P-value* (n=118) (n=60) 8-week TI, n (%) 47 (39.8) 9 (15.0) 6 units of RBCs/8 weeks) and baseline IPSS risk score (Low or Intermediate-1) References on slide 52 8-week TI = proportion of patients without any RBC transfusion for at least eight consecutive weeks since entry to the trial 14
Continuous sustained TI with imetelstat treatment 83% of imetelstat 8-week TI responders had a single continuous TI period * * * Pre-treatment hemoglobin was 6.2 g/dL with transfusion burden of 5 units/8 weeks before study start; on-study hemoglobin was
Median duration of 8-week TI* Highly statistically significant and clinically meaningful durability of TI 1 100% Imetelstat Placebo HR P-value* Percentage of patients with 8-week TI 0.9 90% 0.8 80% Median TI duration (weeks)* 51.6 13.3 0.23
Met key secondary endpoint (24-week TI) Highly statistically significant and clinically meaningful improvement in 24-week TI Median TI duration+ for imetelstat patients who achieved 24-week TI was 80.0 weeks (51.6, NE) Approximately 70% of patients treated with imetelstat who achieved 8-week TI, Imetelstat Placebo went on to achieve 24-week TI P-value* (n=118) (n=60) 24-week TI, n (%) 33 (28.0) 2 (3.3) 6 units of RBCs/8 weeks) and baseline IPSS risk score (Low or Intermediate-1) + Kaplan-Meier estimates of duration of RBC TI References on slide 52 17 24-week TI = proportion of patients without any RBC transfusion for at least 24 consecutive weeks since entry to the trial ; NE = not estimable
Transfusion independence over longer periods Increasing magnitude of benefit for imetelstat vs. placebo with longer time intervals P
Mean change in hemoglobin levels for all imetelstat patients Highly statistically significant increase in hemoglobin levels over time for imetelstat patients 5 8-week TI Imetelstat Placebo P
Mean change in RBC transfusion units over time Statistically significant decrease in number of RBC units transfused 1 P=0.042 Mean Change in RBC Transfusion 0 -1 (units; +/- SE) -2 -3 -4 -5 1 9 17 25 33 41 49 57 65 73 81 89 97 Weeks Imetelstat Placebo Number of patients Imetelstat 115 104 95 76 60 55 45 43 33 26 22 14 10 Placebo 58 53 48 32 27 22 15 14 8 5 5 5 4 P-value is based on a mixed model for repeated measures with change in RBC transfusion as the dependent variable, week, stratification factors, prior transfusion burden, and treatment arm as the independent variables with autoregressive moving average (ARMA(1,1)) covariance structure. References on slide 52 20 NOTE: graph starts at Week 1-8 with the number of the patients with transfusion follow-up data available at least eight weeks on study for imetelstat and placebo arms
Secondary endpoint: hematologic improvement-erythroid (HI-E) Highly statistically significant HI-E using updated IWG 2018 criteria Most recent HI-E criteria (IWG 2018) puts greater emphasis on durability by measuring response at >16 weeks Imetelstat Placebo P-value * (n=118) (n=60) HI-E per IWG 2018 , n (%) 50 (42.4) 8 (13.3)
Key lower risk MDS subtypes analyses Statistically significant 8-week TI rates (p6 units of RBCs/8 weeks) and baseline IPSS risk score (Low or Intermediate-1) ^ One patient on imetelstat arm missing RS category 22 References on slide 52
Primary (8-week TI) and secondary endpoint (24-week TI) by RS status RS+ and RS- demonstrate statistically significant improvement in both 8-week and 24-week TI RS+ RS- 60 P=0.016 Percentage of patients (%) 50 45.2% P=0.003 P=0.038 40 32.9% 31.8% P=0.019 30 18.9% 20.5% 20 8.7% 10 5.4% Imetelstat 0% Placebo 0 8-week 8-week TI n(%) TI rate, 24-week 24-week TI n (%) TI rate, 8-week 8-week TI rate,TIn(%) 24-week 24-week TI n (%) TI rate, RS+ RS- Imetelstat Placebo Imetelstat Placebo P-value* P-value* (n=73) (n=37) (n=44) (n=23) 8-week TI responders, n 33 7 14 2 Median duration of TI *, weeks (95% CI) 46.9 (25.9, 83.9) 16.9 (8.0, 24.9) 0.035 51.6 (11.9, NE) 11.2 (10.1, NE) 0.062 24-week TI responders, n 24 2 9 0 Median duration of TI *, weeks (95% CI) 80.0 (41.6, NE) NE (24.9, NE) 0.808 122.9 (25.0, NE) NE (NE, NE) NE * Kaplan-Meier estimates of duration of RBC TI; 8-week/24-week TI Responder Analysis Set; P-value for TI rate is based on Cochran Mantel Haenszel test stratified for prior RBC transfusion burden (≤6 units or >6 units of RBCs/8 weeks) and baseline IPSS risk score (Low or intermediate-1); P-value for duration of TI is based on stratified log-rank test. 23 References on slide 52
Molecular data in imetelstat treated patients >50% VAF decrease in SF3B1, TET2, DNMT3A, ASXL1 mutations Imetelstat Placebo 45.0 Percentage of patients with ≥50% VAF reduction 40.0% * P=NS 40.0 34.3% * P=NS 35.0 29.5% * P=0.001 30.0 25.0 20.0 16.7% 15.0 11.1% * P=NS 10.0 8.3% 2.6% 5.0 0.0% 0.0 23/78 1/38 12/35 1/12 2/18 0/8 4/10 1/6 SF3B1 TET2 DNMT3A ASXL1 Note: Ratios underneath the bars represent the number of patients with ≥50% variant allele frequency (VAF) reduction as numerator and the total number of patients with detectable assessment (≥5% VAF) in specified mutation at baseline and any post baseline mutation assessment as denominator. * P-value is based on Cochran Mantel Haenszel test stratified for prior RBC transfusion burden (≤6 units or >6 units of RBCs/8 weeks) and baseline IPSS risk score (Low or Intermediate-1) References on slide 52 24 VAF = variant allele frequency; NS = not significant
Efficacy results summary • Trial demonstrated highly statistically significant (p
Top-Line Safety Results Safety results consistent with prior imetelstat clinical experience with no new safety signals 26
Non-hematologic adverse events occurring in ≥10% of patients+ generally low grade and consistent with prior imetelstat clinical trials Imetelstat Placebo AE, n (%) (n=118) (n=59) All Grades Grade 3/4 All Grades Grade 3/4 Asthenia 22 (18.6) 0 8 (13.6) 0 COVID-19* 21 (17.8) 2 (1.7) 9 (15.2) 3 (5.1) Peripheral edema 13 (11.0) 0 8 (13.6) 0 Headache 15 (12.7) 1 (0.8) 3 (5.1) 0 Diarrhea 14 (11.9) 1 (0.8) 7 (11.9) 1 (1.7) Alanine aminotransferase increased 14 (11.9) 3 (2.5) 4 (6.8) 2 (3.4) Hyperbilirubinemia 11 (9.3) 1 (0.8) 6 (10.2) 1 (1.7) Constipation 9 (7.6) 0 7 (11.9) 0 Pyrexia 9 (7.6) 2 (1.7) 7 (11.9) 0 + On either imetelstat or placebo arms * Includes COVID-19, asymptomatic COVID-19, COVID-19 pneumonia References on slide 52 27 AE= adverse event
Treatment emergent liver function test (LFT) lab abnormalities No cases of Hy’s Law or Drug-Induced Liver Injury observed • Grade 3 elevations on imetelstat were short in duration (median < 2 weeks)Placebo Imetelstat and more than 80% LFT resolved to Grade Lab Abnormality, n (%) 2 or lower within 4 weeks (n=118) (n=59) All Grades Grade 3 All Grades Grade 3 Alanine Aminotransferase (ALT*) 46 (39.3) 4 (3.4) 22 (37.3) 3 (5.1) Alkaline Phosphatase (ALP) 53 (44.9) 0 7 (11.9) 0 Aspartate Aminotransferase (AST) 57 (48.3) 1 (0.8) 13 (22.0) 1 (1.7) Bilirubin 46 (39.0) 1 (0.8) 23 (39.0) 1 (1.7) * n=117 for ALT imetelstat patients References on slide 52 28
Consistent with prior imetelstat clinical trials, the most frequent AEs are hematologic Imetelstat Placebo AE, n (%) (n=118) (n=59) All Grades Grade 3/4 All Grades Grade 3/4 Thrombocytopenia 89 (75.4) 73 (61.9) 6 (10.2) 5 (8.5) Neutropenia 87 (73.7) 80 (67.8) 4 (6.8) 2 (3.4) Anemia 24 (20.3) 23 (19.5) 6 (10.2) 4 (6.8) Leukopenia 12 (10.2) 9 (7.6) 1 (1.7) 0 Clinical consequences from cytopenias are similar in imetelstat and placebo treated patients Imetelstat Placebo Event, n (%) (n=118) (n=59) Grade ≥3 Bleeding Events * 3 (2.5) 1 (1.7) Grade ≥3 Infections+ 13 (11.0) 8 (13.6) Grade 3 Febrile Neutropenia ** 1 (0.8) 0 * No ≥Grade 3 bleeding events in the setting of Grade 3/4 thrombocytopenia; on imetelstat: two patients with Grade 4 gastrointestinal bleeding, unrelated and resolved and one Grade 3 hematuria, unrelated and resolved. + On imetelstat: three patients with Grade 3/4 infections in setting of Grade 3/4 neutropenia; all three were sepsis and resolved with only one considered related 29 ** Occurred at day 33, lasted 8 days; assessed by investigator as possibly related to imetelstat; patient subsequently achieved TI >40 weeks and remains on treatment at data cut-off References on slide 52
Imetelstat related cytopenias are manageable and reversible with majority being resolved within
Plans for Regulatory Submissions Lower Risk MDS 31
Top-line results supportive of regulatory submissions On track for 2023 regulatory submissions • Plans on target for regulatory submissions ◦ Mid-2023 for U.S. New Drug Application (NDA) ◦ 2H 2023 for EU Marketing Authorization Application (MAA) • Fast Track designation in lower risk MDS enables rolling submission of U.S. NDA and eligibility for priority review ◦ Rolling submission of NDA initiated • Pre-NDA meeting conducted ◦ No change to submission plans Top-line results supportive of regulatory submissions 32
Unmet Needs and Market Opportunities for Imetelstat Lower Risk MDS 33
Expected broad imetelstat opportunity in lower risk MDS • ~70% of MDS patients are classified MDS: ~37,000* patients diagnosed in the as lower risk US and Major European Markets in 2021 • Majority of the patients HR MDS Lower Risk MDS (~70%) with symptomatic anemia are treated with ESAs and most will fail Complex presentations Symptomatic Anemia (~90%) treatment in ~2 years 1st Line ESA Eligible (~90%) ESA Ineligible (~10%) • Lower risk MDS represents a significant opportunity for imetelstat 2nd Line or later ESA Failed + ESA Ineligible Frontline ESA ineligible RS+ (~25%) RS- (~75%) ESA-failed, RS+ Imetelstat opportunity ESA-failed, RS- * Includes ~10% of patients who may present with del(5q) References on slide 52 HR MDS = higher risk myelodysplastic syndromes; ESA Ineligible = serum EPO >500 mU/mL 34
Imetelstat expected profile at planned launch received favorably by practicing hematologists in both U.S. and key European markets Key aspects of imetelstat that resonated most strongly with hematologists in both the community and academic settings Durability of Potential transfusion for disease independence modification Broad clinical efficacy results Novel MOA, across patient subtypes non-overlapping with (RS+ and RS-) current treatments MOA = mechanism of action References on slide 52 35
Imetelstat potential peak market opportunity in lower risk MDS ~$1.2 billion in 2030 1L ESA Ineligible Patients 3,700 Key addressable patient segments in lower risk MDS for ESA Relapsed/Refractory RS- Patients 22,000 imetelstat* ESA Relapsed/Refractory RS+ Patients 7,000 Imetelstat has the potential to become part of the standard of care in lower risk MDS * Estimated for 2030 based on DRG MDS Landscape and Forecast syndicated data report 2021 and 2022 References on slide 52 1L = first line 36
Commercial Preparedness & Go-to-Market Strategy Key Focus Areas Set up and optimize Hire senior commercial Develop integrated long-lead time supply leadership and team clinical and economic chain and product imetelstat value distribution activities proposition Engage across broad Evolve organization to stakeholder base commercial entity 37
Imetelstat in Relapsed/Refractory MF Evaluating potential improved survival 38
Median OS in IMbark Phase 2 Compares Favorably to Historical Controls ClincialTrials.gov (NCT02426086) Improvement in overall Imetelstat 9.4 mg/kg survival (OS) observed for JAKi relapsed/refractory MF patients in IMbark Phase 2 • 14 – 16 mos median OS for 29.9 mos historical controls for JAKi relapsed/refractory MF patients • 29.9 mos median OS in imetelstat 9.4 mg/kg arm References on slide 52 39
Median OS More than Double Compared to BAT Treatment in RWD Study Evaluating imetelstat vs. BAT in Evaluating imetelstat IMbark Phase 2 data vs.world compared to real BATdata (RWD) from a closely-matched JAKi relapsed/refractory MF in JAKi cohort relapsed/refractory of patients MFwho had discontinued ruxolitinib at the Moffitt Cancer Center and were subsequently treated with best available therapy (BAT) • Improvement in overall survival and • Improvement in overall survival and lower risk of lower risk of death for imetelstat vs. death forRWD BAT vs.vs. imetelstat Imetelstat 9.4 mg/kg BAT in RWD study BAT in RWD study ◦ Imetelstat: 33.8 mos median OS - Imetelstat: 33.8 mos median OS - BAT RWD: 12.0 mos median OS BAT Moffitt Imetelstat 9.4 mg/kg ◦ BAT RWD: 12.0 mos median OS - ◦ 65% lower risk of death with imetelstat Survival Probability 65% lower risk of death with 33.8 imetelstat mos compared to BAT from RWD compared to BAT from RWD • OS improvement 12.0 mos and lower risk of death for • OS improvement and lower risk of imetelstat vs. BAT support IMpactMF Phase 3 trial death for imetelstat vs. BAT support design IMpactMF Phase 3 trial design • Acknowledging Same dose and schedule being used in IMpactMF trialPhase the limitations of such comparative analyses between RWD and clinical 3 trialthe favorable overall survival (OS) of imetelstat treatment suggested data, we believe • Same dose and schedule being used in by these comparative analyses in this very poor prognosis patient population warrants IMpactMF Phase 3 trial further evaluation. References on slide 52 40
Manageable Imetelstat Safety Results in IMbark Phase 2 Limited clinical consequences of 9.4 mg/kg (n=59) reversible, on target cytopenias n (%) All Grades Grade ≥ 3 ClincialTrials.gov (NCT02426086) • Thrombocytopenia and neutropenia Hematologic (≥10% in either arm)§ characterization: Thrombocytopenia 29 (49) 24 (41) ◦ Short time to onset: Median 9 weeks Anemia 26 (44) 23 (39) (~3 cycles) Neutropenia 21 (36) 19 (32) ◦ Short duration: Median 70% within 4 weeks* Nausea 20 (34) 2 (3) ◦ Manageable with dose hold and modifications Diarrhea 18 (31) 0 Fatigue 16 (27) 4 (7) • Limited clinical consequences: Dyspnea 14 (24) 3 (5) ◦ 2% Grade 3 febrile neutropenia Abdominal Pain 14 (24) 3 (5) ◦ 5% Grade 3/4 hemorrhagic events Asthenia 14 (24) 6 (10) ◦ 10% Grade 3/4 infections §Treatment emergent, per reported AEs (not laboratory Pyrexia 13 (22) 3 (5) values). Frequency of reported Grade 3/4 hematologic adverse events were Edema peripheral 11 (19) 0 consistent with cytopenias reported through lab values. *Reversible to Grade 2 or lower. References on slide 52 41
Phase 3 IMpactMF Trial in Relapsed/Refractory MF ClincialTrials.gov (NCT0457615) Actively enrolling patients now Imetelstat Primary Endpoint: Intermediate-2 9.4mg/kg every 3 weeks • Overall survival (OS) or High-Risk MF (n ~214) Relapsed/Refractory to 2:1 Key Secondary Endpoints: JAK inhibitors (JAKi) • Symptom response Best Available • Spleen response (n=320) Therapy (BAT) • Patient Reported Outcomes (PROs) (n ~106) References on slide 52 42
First and Only Phase 3 Trial in the U.S. in MF with OS as Primary Endpoint Actively enrolling global trial • Sites planned across North America, South America, Europe, Australia and Asia ClincialTrials.gov (NCT0457615) Planned analyses Primary Endpoint: • Interim Analysis expected in 2024 when ~35% of the planned enrolled patients have died; alpha spend ~0.01 • Overall survival (OS) • Final Analysis expected in 2025 when >50% of the planned enrolled patients have died Key Secondary Endpoints: Statistically well-powered trial • Symptom response • Designed with >85% power to detect a 40% reduction in the risk of death in the imetelstat arm compared to BAT (hazard • Spleen response ratio=0.60; one-sided alpha=0.025) • Patient Reported Outcomes (PROs) • Conservative powering assumptions - Median OS: 14 mos for BAT vs. 23 mos for imetelstat References on slide 52 43
Unmet Needs and Market Opportunities for Imetelstat Relapsed/Refractory MF 44
Expected MF Market Evolution and Imetelstat Opportunity* We expect the future MF market to expand significantly with potential approvals of JAKi-based combination regimens and treatments that address anemia in MF patients. Agents in development today have primary endpoints focused on addressing spleen, symptoms and anemia Int-2/High Risk MF Patients Treated with JAK Inhibitors ~75% discontinuation rate after 5 years Unresponsive to JAK Inhibitors Almost all JAKi- median OS ~14 – 16 months per literature reviews treated patients expected to become NO APPROVED THERAPY unresponsive to JAKis and eligible for Expected Imetelstat Addressable Patient Population imetelstat ~18,000 patients (US/EU5) USD ~$1.8 Billion in Potential Peak Market Opportunity Across US & Key European Markets** *Projections in 2030 in Int-2/HR MF **Company projections: based on treated prevalence estimates for imetelstat eligible patient populations in Int-2/HR MF (2030); DRG syndicated data, US/G5 payor research and Geron analysis using assumptions for a) expected target product profile at launch, b) obtaining regulatory approvals and favorable reimbursement in US and key European markets, c) duration of treatment and d) potential market penetration; Company estimate does not include Int-1 & platelets
Financial Overview 46
Financial Resources to Support Potential Commercial Launch of Imetelstat ~$173M ~$213M ~$60M Financial resources As of Net proceeds Proceeds from expected to support December 31, from Jan 2023 warrant exercises 2022 financing in Jan/Feb 2023 projected level of operations through third quarter 2025^ ^ Based on current operating plan and expectations regarding the timing of regulatory submissions, and potential acceptance and approval of planned NDA by the FDA in the U.S. for the use of imetelstat in adult patients with lower risk MDS and the potential subsequent commercialization in 1H 2024. 47
Investment Thesis 48
Exploring the broad potential of imetelstat and telomerase inhibition Indications Discovery Preclinical Phase 1 Phase 2 Phase 3 LR MDS IMerge Single Agent R/R MF Single Agent IMpactMF Frontline MF Combination Therapy IMproveMF R/R AML & HR MDS IMpress Single Agent R/R AML Combination Therapy TELOMERE* Lymphoid Malignancies Ongoing; Company Sponsored Next Generation TI Program Planned; Investigator Led * To follow single agent data from IMpress LR MDS = lower risk myelodysplastic syndromes; R/R MF = relapsed/refractory myelofibrosis; MF = myelofibrosis; R/R AML = relapsed/refractory acute myeloid leukemia; HR MDS = higher risk myelodysplastic syndromes; TI = telomerase inhibitor 49
Imetelstat - a highly differentiated drug • Unique Mechanism of Action • Broad and durable TI in LR MDS demonstrated in IMerge Phase 3 • Potential OS readout in R/R MF • Evidence for potential disease modification Multiple upcoming catalysts expected Key Takeaways • NDA submission in LR MDS – mid-2023 • MAA submission in LR MDS – 2H 2023 • Interim analysis in R/R MF - 2024 Potential commercial future • Multi-billion dollar market potential • U.S. LR MDS launch planned for first half 2024* • EU planning underway * If the NDA is accepted for filing and imetelstat is approved for commercialization by the FDA References on slide 52 50
Thank you! Contact: Investor Relations info@geron.com
References Slide # Reference Slide # Reference Slide # Reference Slide # Reference 3, 4, IMerge Phase 3 TLR reported Jan 4, 2023 35 KOL Interviews, Advisory Boards, Geron Physician 45 MF patient numbers: Geron Physician Market Research 60 Hu et al, ASH 2019 9-30 Market Research (US/EU5); stimuli included IMerge December 2021, US Hematologist Market survey Phase 2 LR MDS data and expected target product (n=100); company projections in 2030 profile at launch Market potential: Company projections in 2030 based on treated prevalence estimates for imetelstat eligible patient populations in Int-2/HR MF; DRG syndicated data; Payor research (US/EU5) and Geron analysis using assumptions for a) expected target product profile at launch, b) obtaining regulatory approvals and favorable reimbursement in US and key European markets, c) duration of treatment and d) potential market penetration; for MF, does not include Int-1 & platelets
Appendix 1 Strong Phase 2 evidence for potential disease modification 53
Strong Evidence of Imetelstat Disease Modification in Phase 2 Studies Killing of Recovery of Bone Reduced Telomerase Activity Malignant Stem and Marrow and Blood Clinical Benefits Progenitor Cells Cell Production Correlated with Depletion of mutated and Significant hemoglobin Depth and clinical benefits, cytogenetically abnormal rise in 75% of responders durability of MDS confirms MOA malignant cells correlated transfusion with clinical benefits independence Correlated with Depletion of cells with Improvements clinical benefits, key driver mutations and in bone marrow Improved MF confirms MOA abnormal cytogenetics fibrosis correlated overall survival correlated with improved OS to improved OS and symptoms References on slide 52 54 MOA = mechanism of action; OS = overall survival
Strong Evidence of Disease Modification Potential ClinicalTrials.gov (NCT02598661) Imetelstat results in reduction of malignant clones in IMerge Phase 2 Change in VAF of Percent SF3B1 VAF Reduction Imetelstat Reduces Abnormal A SF3B1 Mutations B vs. Longest TI Duration C Cytogenetic Clones 100% 95% 95% 100% Cytogenetic Clones 75% 80% % of Abnormal Reduction (%) SF3B1 VAF VAF (%) 60% 45% 40% 25% 20% 5% 5% 5% 0% Baseline ~24 wks ~48 wks ~72 wks Longest Transfusion- 47,XX,+8 47,XY,+8 46,XX,Dup/Tri/Qtp(9)(P13P24) Free Interval (Weeks) Greater SF3B1 variant allele frequency Patients with abnormal cytogenetics at Reduction of SF3B1 mutation burden in LR reduction (VAF) correlates baseline have reduction of their clones and MDS patients treated with imetelstat with longer duration of TI long (>1year) TI; 2 of 3 patients achieved partial cytogenetic response References on slide 52 VAF = variant allele frequency; TI = transfusion independence 55
Strong Evidence of Disease Modification Potential ClincialTrials.gov (NCT02426086) Improved bone marrow fibrosis correlated to improved survival in IMbark Phase 2 Significant Dose-Dependent Fibrosis Longer Median OS and Higher Survival Rate Improvement with Imetelstat Treatment in Patients with Improved Fibrosis 1.0 % Achieved BM Fibrosis 0.9 CENSORED Yes Improvement 0.8 No Survival Probability 0.7 0.6 0.5 41% 0.4 BM Fibrosis Median OS HR Improvement (months) (95% CI) (95% CI) 0.3 31.6 YES 20% 0.2 (23.6, NE) 0.54 24.6 (0.23, 1.29) NO 0.1 (18.4, NE) 0.0 4.7 mg/kg 9.4 mg/kg 0 6 12 18 24 30 36 Time on Study (months) References on slide 52 OS = overall survival; BM = bone marrow; HR = hazard ratio; CI = confidence interval; NE = not evaluable 56
Strong Evidence of Disease Modification Potential ClincialTrials.gov (NCT02426086) Reduction in key MF driver mutations correlated to improved survival in IMbark Phase 2 Significant Dose-Dependent ≥20% VAF Longer Median OS and Higher Survival Rate in Patients Reduction with Imetelstat Treatment Who Achieved ≥ 20% VAF Reduction p=0.0321 ≥20% VAF Median OS HR Reduction (months) (95% CI) (95% CI) 31.6 YES (21.5, –) 0.512 % Achieved >=20% Reduction of VAF 22.8 (0.238, 1.100) NO (17.1, 31.6) Survival Probability 46% 17% 4.7 mg/kg 9.4 mg/kg Time on Study (months) References on slide 52 VAF = variant allele frequency; OS = overall survival; HR = hazard ratio; CI = confidence interval 57
Strong Evidence of Disease Modification Potential ClincialTrials.gov (NCT02426086) Malignant clones reduced in imetelstat-treated patients in IMbark Phase 2 Dose-Dependent Complete Elimination of Mutation Burden from Multiple Driver- and Non-Driver Genes 9.4 9.4 mg/kg mg/kg 4.7 4.7 mg/kg mg/kg Completely eliminated JAK2 >=20% reduction CALR >=10% reduction MPL No change ASXL1 Acquired new clone EZH2 >=20% increase IDH2 SRSF2 >=10% increase SF3B1 U2AF1 • Mutation status and variant DNMT3A allele frequency (VAF) were TET2 evaluated by next- CBL generation sequencing KRAS (NGS) using Illumina TruSight Myeloid NRAS Sequencing Panel of TP53 54-genes GATA2 BCORL1 • Lower limit detection is 5% ETV6 and 2% for well documented KIT hotspots PHF6 • 49 pts had matched pre- and RUNX1 at least a post-imetelstat STAG2 treatment NGS data References on slide 52 58
Appendix 2 Pipeline expansion programs 59
Phase 1 IMproveMF Study Evaluating Combination Therapy in Frontline MF Exploring the potential for disease modification with Imetelstat earlier in MF disease setting Study Started Preclinical data for ruxolitinib followed by imetelstat: • Additive inhibitory effect on MF stem cells in vivo May 2022 • Synergistically depletes stem cells from MF patients (PDX model) Rationale Part 1: Dose Finding Part 2: Dose Confirmation & Expansion (n = up to 20) (n = ~ 20) Ruxolitinib Imetelstat Ruxolitinib Frontline MF + Phase 2 Dose + DIPSS int1/int2/HR Imetelstat Imetelstat Selected single arm open label single arm open label • Objective: Identify recommended • Objective: Confirm safety of doses Phase 2 doses and evaluate efficacy • Primary Wk 24 Endpoint: Safety • Primary Wk 24 Endpoints: Safety, TSS • Other Wk 24 Endpoints: TSS, SVR35, Fibrosis • Other Wk 24 Endpoints: TSS, SVR35, Fibrosis References on slide 52 RP2D = Recommended Phase 2 Dose; DIPSS = dynamic international prognostic scoring system; int1 = intermediate-1; int2 = intermediate-2; HR = high risk; TSS = total symptom score; SVR35 = spleen volume reduction > 35%; TI = transfusion independence; IWG-MRT = international working group-myeloproliferative neoplasms research and treatment 60
IMpress – Planned Phase 2 Investigator-Led Study of Single Agent Imetelstat in Post-HMA Relapsed/Refractory AML Lead PrincipaI Investigator: Dr. Uwe Platzbecker, University Hospital, Leipzig, Germany In preclinical models, imetelstat: Planned First Site • Prevented expansion of human AML leukemic stem cells (PDX model) Opening: • Prolonged survival of AML PDX mice 2023 Rationale Open-label, Single-arm Multicenter (n = ~ 45) Objective: Evaluate efficacy R/R/Intolerant Imetelstat HR MDS and AML Endpoint: Overall Response Rate per 7.5mg/kg I.V. IWG 2018 criteria (MDS) and the criteria Post HMA of the European LeukemiaNet (AML) References on slide 52 PDX = patient-derived xenografts; R/R = relapsed or refractory; HR MDS = intermediate-2 or high risk MDS per International Prognostic Scoring System; AML = acute myeloid leukemia; HMA = hypomethylating agent; IWG 2018 = international working group 2018 criteria for hematologic response 61
The Tisch TELOMERE – Pending Phase 1/2 Investigator-Led Study of Imetelstat in Cancer Institute Combination with Venetoclax or Azacitidine in Relapsed/Refractory AML Lead Principal Investigator: Dr. John Mascarenhas, Mt. Sinai Hospital, New York, New York In preclinical models, imetelstat + venetoclax (Ven): • Synergistically induced apoptosis in primary AML blasts ex vivo Study Start Expected: • Enhanced survival with potential cure in AML xenograft model Pending In preclinical models, imetelstat + azacitidine (Aza): IMpress Data Rationale • Synergistically induced apoptosis in AML cell lines Phase 1 Dose Finding (n = up to 20) Phase 2 SIMON 2 Stage Design (n = ~ 50) STAGE 1 STAGE 2 STABLE DISEASE Imetelstat + Aza PD Imetelstat + Aza Imetelstat + Aza Sufficient number of R/R AML responders needed in each arm otherwise, Post Aza and/or Ven the arm is discontinued Imetelstat + Ven PD Imetelstat + Ven Imetelstat + Ven STABLE DISEASE • Objective: Identify recommended Phase 2 dose for each combination • Objective: Evaluate efficacy for each combination • Endpoint: Safety for each combination • Endpoints: Overall Response Rate for each combination References on slide 52 R/R = relapsed or refractory; AML = acute myeloid leukemia; PD = progressive disease 62
Ongoing and Planned Preclinical Experiments to Define the Role of Imetelstat in Lymphoid Malignancies Lead Principal Investigator: Dr. Swaminathan Iyer, MD Anderson Cancer Center, Houston, Texas T-Cell lymphoma cell lines in vitro have shown: • Short telomere length (Sezary syndrome; transformed mycosis fungoides) • High telomerase activity (cut. anaplastic large-cell Further experiments of lymphoma; mycosis fungoides) imetelstat in lymphoid malignancies expected In Cutaneous T-Cell lymphoma cell lines in vitro: Rationale • Telomerase overexpression increased T-Cell proliferation • RNAi inhibition of telomerase decreased T-Cell proliferation In vitro assays in cell lines and patient-derived materials In vivo mouse studies Studies Being (blood, etc.) from T and B-cell lymphomas in T and B-cell Conducted at MD • Apoptosis (cell death) assays lymphoma models • Colony forming cell (CFCs) assays Anderson Cancer • Cytokine assays Center • TA, TL, hTERT assays References on slide 52 TA = telomerase activity; TL = telomere length; hTERT = human telomerase reverse transcriptase 63
Next Generation Telomerase Inhibitor Program Update Status • Expect completion of Program goal current discovery effort in 2023, at which time Discover and develop novel small molecules based on chemistry Geron plans to potentially advance platforms proprietary to Geron that bind to the active site of the any lead compounds into the next step of telomerase molecule and directly inhibit telomerase activity discovery research. • If successful, these efforts would permit initiation Aspirational profile of IND-enabling nonclinical studies. of a lead compound candidate Oral High potency High potential for delivery & selectivity combinability 64
You can also read