Emerging Treatment Options for Myelodysplastic Syndromes

Similar documents
Emerging Treatment Options for Myelodysplastic Syndromes

Emerging Treatment Options for Myelodysplastic Syndromes

Myelodysplastic syndromes post ASH Dominik Selleslag AZ Sint-Jan Brugge

Treatment of low risk MDS

Low Risk MDS Scoring System. Prognosis in Low Risk MDS. LR-PSS Validation 9/19/2012

Myelodysplastic Syndromes. Post-ASH meeting 2014 Marie-Christiane Vekemans

Novità nelle MDS. Matteo G Della Porta. Cancer Center IRCCS Humanitas Research Hospital & Humanitas University Rozzano Milano, Italy

New and Emerging Strategies in the Treatment of Patients with Higher risk Myelodysplastic Syndromes (MDS)

Treating Higher-Risk MDS. Case presentation. Defining higher risk MDS. IPSS WHO IPSS: WPSS MD Anderson PSS

Molecular Genetic Testing to Predict Response to Therapy in MDS

MDS-004 Study: REVLIMID (lenalidomide) versus Placebo in Myelodysplastic Syndromes with Deletion (5q) Abnormality

Understanding & Treating Myelodysplastic Syndrome (MDS)

MDS FDA-approved Drugs

Myelodysplastic syndromes: 2018 update on diagnosis, risk-stratification and management

Myelodysplastic syndromes Impact of Biology. Lionel Adès Hopital Saint Louis Groupe Francophone des SMD. Épidémiologie

Overview. Myelodysplastic Syndromes: What s on the Horizon? Molecular Mutations in MDS. Refining Risk Models. Incorporating Mutational Data

Refining Prognosis. Overview. Low Blood Counts. Low Blood Counts. High Risk MDS and Novel Therapy: What s on the Horizon? 3/2/2016

Myelodysplastic Syndrome: Let s build a definition

MDS 101. What is bone marrow? Myelodysplastic Syndrome: Let s build a definition. Dysplastic? Syndrome? 5/22/2014. What does bone marrow do?

La lenalidomide: meccanismo d azione e risultati terapeutici. F. Ferrara

Let s Look at Our Blood

Disclosure Slide. Research Support: Onconova Therapeutics, Celgene

Myelodysplastic syndromes

Acute Myeloid Leukemia

The Changing Face of MDS: Advances in Treatment

A Phase II Study of the Combination of Oral Rigosertib and Azacitidine in Patients with Myelodysplastic Syndromes (MDS)

Pathogenesis and management of CMML

MYELODYSPLASTIC SYNDROMES: A diagnosis often missed

[ NASDAQ: MEIP ] Analyst & Investor Event December 8, 2014

Myelodysplastic Syndromes: Understanding your diagnosis and current and emerging treatments

Table 1: biological tests in SMD

What you need to know about MDS. The Myelodysplastic Syndromes. Stuart Goldberg MD

Your Speaker. Outline 10/2/2017. Myelodysplastic Syndromes and Myeloid Neoplasms. Introduction and classifications Epidemiology Presentation Workup

Outline. Case Study 5/17/2010. Treating Lower-Risk Myelodysplastic Syndrome (MDS) Tapan M. Kadia, MD Department of Leukemia MD Anderson Cancer Center

Combination of Oral Rigosertib and Injectable Azacitidine in Patients with Myelodysplastic Syndromes (MDS)

IPSS Modified 7/27/2011. WHO-Based Prognostic Scoring System (WPSS)

Management of low and high risk MDS

Acute Myeloid Leukemia Progress at last

MDS: Who gets it and how is it diagnosed?

MYELODYSPLASTIC SYNDROME. Vivienne Fairley Clinical Nurse Specialist Sheffield

INTRODUCTION TO CYTOGENETICS AND MOLECULAR TESTING IN MDS

New treatment strategies in myelodysplastic syndromes and acute myeloid leukemia van der Helm, Lidia Henrieke

AML IN OLDER PATIENTS Whenever possible, intensive induction therapy should be considered

APPROACH TO MYELODYSPLASTIC SYNDROMES IN THE ERA OF PRECISION MEDICINE

Overview of Aplastic Anemia. Overview of Aplastic Anemia. Epidemiology of aplastic anemia. Normal hematopoiesis 10/6/2017

ASBMT MDS/MPN UPDATE

Summary of Key AML Abstracts Presented at the European Hematology Association (EHA) June 22-25, 2017 Madrid, Spain

Myelodysplastic syndrome. Jeanne Palmer, MD Mayo Clinic, Arizona

MDS overview 전남대학교김여경

ACCME/Disclosures. History. Hematopathology Specialty Conference Case #4 4/13/2016

Piper Jaffray Healthcare Conference

Maintaining Long-Term Efficacy in the Elderly MDS Patient with Poor Performance Status

CREDIT DESIGNATION STATEMENT

Mutational Impact on Diagnostic and Prognostic Evaluation of MDS

When Cancer Looks Like Something Else: How Does Mutational Profiling Inform the Diagnosis of Myelodysplasia?

Dr Kavita Raj Consultant Haematologist Guys and St Thomas Hospital

Corporate Presentation. January 2019

DISCLOSURE Luca Malcovati, MD. No financial relationships to disclose

Current guidelines for management of INT- 2/high risk MDS

Myelodysplastic Syndrome

ASBMT MDS/MPN Update Sunil Abhyankar, MD

Needham Healthcare Conference. John Scarlett, M.D. President & CEO, Geron Corporation March 27, 2018

Myelodysplastic Syndromes (MDS) Enhancing the Nurses Role in Management

About Myelodysplastic Syndromes

Non-transplant Therapy for MDS. Bart Scott, MD Associate Member, FHCRC Associate Professor, UWMC

Management of Myelodysplastic Syndromes

Corporate Presentation January John Scarlett, M.D., President & CEO Olivia Bloom, EVP Finance & CFO Anna Krassowska, Ph.D., Investor Relations

Molecular profiling in confirming the diagnosis of early myelodysplastic syndrome

Blast transformation in chronic myelomonocytic leukemia: Risk factors, genetic features, survival, and treatment outcome

Myelodysplastic Syndromes:

Session II: Summary. Robert P Hasserjian, MD Professor of Pathology Massachusetts General Hospital and Harvard Medical School

Chronic myelomonocytic leukemia. Lymphoma Tumor Board. May 26, 2017

AML in elderly. D.Selleslag AZ Sint-Jan Brugge, Belgium 14 December 2013

2/7/2017. Updates in MDS: Overview

EHA 2017 Abstracts: 4 Abstracts ( 1 Oral Presentation, 2 EPosters, 1 Publication)

Myelodysplastic syndromes: revised WHO classification and distinction from non-neoplastic conditions

CHALLENGING CASES PRESENTATION

Kevin Kelly, MD, Phd Acute Myeloid and Lymphoid Leukemias

Guidelines for diagnosis and management of Adult Myelodysplastic Syndromes (MDS)

Published Ahead of Print on April 14, 2016, as doi: /haematol Copyright 2016 Ferrata Storti Foundation.

Myelodysplastic syndromes and the new WHO 2016 classification

IDH1 AND IDH2 MUTATIONS

SWOG ONCOLOGY RESEARCH PROFESSIONAL (ORP) MANUAL LEUKEMIA FORMS CHAPTER 16A REVISED: DECEMBER 2017

Etiology. Definition MYELODYSPLASTIC SYNDROMES. De novo. Secondary MDS (10 years earlier than primary) transformation

Summary of Key AML Abstracts Presented at the American Society of Hematology (ASH) December 2-6, San Diego CA

Use of TPO mimetics for Indications Other Than ITP

Post-ASH 2015 CML - MPN

Myelodysplastic Syndrome. Ryotaro Nakamura, MD Associated Professor Department of Hematology and Hematopoietic Cell Transplantation

Combination Therapies in Higher-risk MDS

Prognostic Scoring Systems for Therapeutic Decision Making in MDS. Peter Greenberg, MD Stanford University Cancer Center Stanford, CA

Scottish Medicines Consortium

Shyamala Navada, MD Icahn School of Medicine at Mount Sinai. 60th ASH Annual Meeting & Exposition 2018: San Diego, CA, USA 1

What is MDS? Epidemiology, Diagnosis, Classification & Risk Stratification

MANAGEMENT OF ADULT PATIENTS WITH MYELODYSPLASTIC SYNDROMES

Myelodyplastic Syndromes Paul J. Shami, M.D.

Myelodysplastic Syndrome. Ryotaro Nakamura, MD Associated Professor Department of Hematology and Hematopoietic Cell Transplantation

29 th Annual Piper Jaffray Healthcare Conference. November 28-29, 2017

Randomized phase 2 study of low-dose decitabine vs low-dose azacitidine in lower-risk MDS and MDS/MPN

Understanding AML Casey O Connell, MD Associate Professor, Jane Anne Nohl Division of Hematology Keck School of Medicine, USC

Treatment of Low-Blast Count AML. Maria Teresa Voso Dipartimento di Biomedicina e Prevenzione Università di Roma Tor Vergata

Transcription:

Emerging Treatment Options for Myelodysplastic Syndromes James K. Mangan, MD, PhD Assistant Professor of Clinical Medicine Abramson Cancer Center, University of Pennsylvania Please note that some of the studies reported in this presentation were presented as an abstract and/or presented at a conference. These data and conclusions should be considered to be preliminary until published in a peer-reviewed journal.

Age Related Incidence of MDS A group of heterogeneous clonal hematopoietic cell neoplasms Overall incidence 5 per 100,000 Most common myeloid neoplasm Peripheral blood cytopenias Variable risk of progression to AML (1 in 3) Rate per 1000,000 population SEER data 2008-2012 and Greenberg, et al 1997 Reproduced with permission, H. Carraway

Making a diagnosis of Myelodysplastic syndromes (MDS) At least one cytopenia: Hb <11 g/dl, or ANC <1500/μL, or Platelets <100 x 10 9 /L MDS decisive criteria: >10% dysplastic cells in 1 or more lineages, or 5-19% blasts, or Abnormal karyotype typical for MDS EXCLUDE other causes of cytopenias and morphological changes: Vitamin B12/folate deficiency HIV or other viral infection Copper deficiency Alcohol abuse Medications (esp. methotrexate, azathioprine, recent chemotherapy) Autoimmune conditions (ITP, Felty syndrome, SLE etc.) Congenital syndromes (Fanconi anemia etc.) Other hematological disorders (aplastic anemia, LGL disorders, MPN etc.) Slide courtesy of R. Komrokji (modified)

CHIP: Clonal Hematopoiesis of Indeterminate Potential Absence of definitive morphologic criteria of a heme malignancy Not meeting diagnostic criteria for MGUS, MBL, or PNH Detection of a recurrent hematologic malignancy-associated somatic molecular mutation at a VAF of 2% (e.g. TET2, DNMT3A, etc.) Prevalence increase with age Approximately 10% of people >65-70 years progression to overt malignancy (0.5-1%/year, similar to MGUS) Higher risk of CAD (HR= 2) and ischemic strokes (HR=2.6) Higher risk of all-cause mortality (HR=1.4) Steensma D et al, Blood, 2015; Jaiswal et al, NEJM, 2015, Genovese G et al, NEJM, 2015 Slide reproduced from the American Society of Hematology

IPSS: First Tool for Risk Stratification of MDS Score Value Prognostic variable 0 0.5 1.0 1.5 2.0 Bone marrow blasts < 5% 5% to 10% -- 11% to 20% 21% to 30% Karyotype* Good Intermediate Poor -- -- Cytopenias 0/1 2/3 -- -- -- Risk Group Risk Score Low 0 Intermediate 1 0.5-1 Intermediate 2 1.5-2 High >/= 2.5 Low risk High risk *Good = normal, -Y, del(5q), del(20q); intermediate = other karyotypic abnormalities; poor = complex ( 3 abnormalities) or chromosome 7 abnormalities. Hb < 10 g/dl; ANC < 1800/µL; platelets < 100,000/µL. Greenberg P, et al. Blood. 1997;89:2079-2088.

IPSS-R: Used to Determine Prognosis/Risk P. Variable 0 0.5 1 1.5 2 3 4 Cytogenetics Very Good Good Intermediate Poor Very Poor BM Blast % 2 >2-<5% 5-10% >10% Hemoglobin 10 8-<10 <8 Platelets 100 50-<100 <50 ANC 0.8 <0.8 IPSS-R: Prognostic Risk Categories/Scores Risk Group Risk Score Very Low 1.5 Low >1.5-3 Intermediate >3-4.5 High >4.5-6 Very High >6 Low risk High risk Greenberg et al. Blood 2012;120:2454-65.

Prognosis: IPSS-R

Somatic mutation in any of the 5 genes (TP53, EZH2, RUNX1, ASLX1, or ETV6) shown in Bejar et al, NEJM 364, 2011 to have prognostic significance independent of the International Prognostic Scoring System (IPSS) identifies patients from that same cohort with shorter overall survival than predicted by the IPSS-R for the 3 lowest IPSS-R risk groups. Bejar R, and Steensma D P Blood 2014;124:2793-2803 2014 by American Society of Hematology

IPSS-Rm ASH abstract 607 2015, Aziz Nazha Et al. 508 patients. Three genes were found to be prognostic: EZH2, p53, SF3B1.

2015 by American Society of Hematology Rafael Bejar et al. Blood 2015;126:907

2014 by American Society of Hematology Bejar R et al. Blood 2014;124:532

Treatment Goals in MDS Cure Improve overall survival Lower the risk of transformation to AML Complete remission, partial remission, stable disease Improve tri-lineage hematopoiesis and function o Decrease infections by resolving neutropenic state/dysfunction o Decrease PRBC/PLT transfusion burden Supportive care o Decrease symptoms that impair quality of life Reproduced with permission, H. Carraway

Therapies for Lower Risk MDS ESAs: Work well in patients with low transfusion Requirements (<2 units PRBCs/month) and EPO level (<500): 74% response rate. Work poorly in patients with high transfusion Requirements and EPO >500: 7% response rate (Hellstrom- Lindberg et al, BJH 120, 2003). Also clearly inferior responses in higher risk disease.

Riva M et al, ASH abstract 2981, 2017.

Low and Int-1 myelodysplastic syndrome Age 18 Arm A 6 cycles Trial Design HOVON89 Randomize Arm B 4 cycles 2 cycles No HI Off treatment 2 cycles No HI Relapse after HI 2 cycles 1 Epo 30,000 IU No HI 2 cycles 1 Epo 60,000 IU Relapse after HI Off treatment 2 cycles Relapse after HI 2 cycles 1 Epo 30,000 IU Relapse after HI No HI 2 cycles 2 cycles Relapse after HI Relapse after HI Off treatment Off treatment 2 cycles 2 cycles Relapse after HI Maintenance cycles Relapse after HI Off treatment Relapse after HI or no HI 2 cycles 1 Epo 30000 IU 2 cycles 1 Epo 30,000 IU Maintenance cycles Epo 30,000 IU Relapse after HI 2 cycles 1 Epo 60,000 IU 2 cycles 1 Epo 60,000 IU Relapse after HI or no HI Maintenance cycles Epo 60,000 IU Relapse after HI Off treatment 2 cycles 1 Epo 60,000 IU G-CSF 2 cycles 1 Epo 60,000 IU G-CSF Relapse after HI or no HI Maintenance cycles Epo 60,000 IU G-CSF Disease progression or baseline transfusion requirements After each cycle: Haematological evaluation of PB should be performed If disease progression or baseline transfusion requirements Off treatment Off treatment 1 For Epo and/or G-CSF reduction in case of HI, see protocol 9.2.2 and 9.2.3. Van DeLoosdrecht et al. ASH 2016; Abs 224

HOVON89 Results: Primary and Secondary Endpoints Hematological Improvement-Erythroid*: 41% 39% and 42% for the pts in arm A and B, respectively (p = 0.45) Hematological Improvement-Erythroid*: non-del5q versus del5q: 34% vs 79% Time-to-HI-E: 3.1 months (median; range 1.6-12.3 ) for both arms Duration of HI-E: 10.6 months (range 1.4 76.1) *According to IWG criteria Van DeLoosdrecht et al. ASH 2016; Abs 224

Results: NGS and Response to Lenalidomide Presence of 2 or more mutations are inversely related to HI-E (p=0.004) Presence of 1 or more splicing factor mutations are inversely related to HI-E (p<0.0001) Of the 7 most frequently mutated genes (i.e.) TET2, ASXL1, DNMT3, ATRX, RUNX1, only SRSF2 (p=0.021) and SF3B1 (p=0.004) are significantly associated with lack of response to (HI-E) Van DeLoosdrecht et al. ASH 2016; Abs 224

Luspatercept PACE-MDS Phase 2 Clinical Trials Overview A Phase 2, multicenter, open-label, 3-month dose-escalation study in adults with lower-risk MDS followed by a 5-year extension study Patient Population Multiple cohorts enrolling low/intermediate-1 risk (IPSS) MDS patients including: Non-transfusion dependent and transfusion dependent patients ESA-naïve and ESA-experienced patients Patients with a range of baseline EPO levels RS+ and non-rs patients Treatment Base Study (N=106) 3 months NCT01749514 Luspatercept 0.125 1.75 mg/kg (base study); 1.0 1.75 mg/kg (extension) SC q3 weeks All patients followed up for 2 months post last dose or early discontinuation Extension Study (N=70) 5 years (ongoing) NCT02268383 Efficacy Endpoints IWG (2006) HI-E: Hb increase 1.5 g/dl for all values over 8 weeks for patients with < 4 units/8 wk and Hb < 10 g/dl 4 RBC unit decrease over 8 weeks for patients with 4 units/8 wk Other Efficacy Endpoints RBC-TI: RBC-transfusion independence 8 weeks (RBC evaluable patients, 2U/8 weeks) Time to/duration of HI-E response EPO: erythropoietin; ESA: erythropoiesis-stimulating agent; HI-E: hematologic improvement erythroid; RS: ring sideroblast 18 Platzbecker U et al. ASH 2017 [Abstract # 2982] Data as of 08 Sept 2017

IWG HI-E and RBC-TI Response Rates by ESA, EPO, RS Status Patients Treated at Dose Levels 0.75 mg/kg Response Rates IWG-HI-E, n/n (%) (N=99) RBC-TI, n/n (%) (N=67) All patients 52/99 (53%) 29/67 (43%) ESA-naïve 28/53 (53%) 17/31 (55%) Prior ESA 24/46 (52%) 12/36 (33%) Baseline EPO <200 U/L RS+ 25/39 (64%) 16/24 (67%) Non-RS 7/13 (54%) 3/7 (43%) Baseline EPO 200-500 U/L RS+ 10/14 (71%) 4/9 (44%) Non-RS 4/8 (50%) 3/5 (60%) RS Status RS+ 40/62 (65%) 22/42 (52%) Non-RS 12/35 (34%) 7/23 (30%) Unknown 0/2 (0%) 0/2 (0%) 67% HI-E in SF3B1 mutated (N= 43)vs 32% in WT (N=31) 48% RBC-TI in SF3B1 mutated (N= 29)vs 36% in WT (N= 22) Platzbecker U et al. ASH 2017 [Abstract # 2982] Data as of 08 Sept 2017

HMAs might improve survival in some CMML patients N died Pre-HMA (N=395) HMA (N=225) HMA vs non-hma (ref) Median survival (95% CI) N died Median survival (95% CI) HR (95% CI) P-value 385 11 [8-13] 200 17 [14-19] 0.72 (0.58-0.91).005 1.0 + Censored 0.8 Survival Probability 0.6 0.4 0.2 0.0 0 6 12 18 24 30 36 42 48 54 60 followup_month ps_time All before 2004 HMA after 2006 Zeidan A et al, Cancer, 2017

Study Design: Phase 1/2 Ruxolitinib CMML Study Phase One 5mg BID 10mg BID 15mg BID Phase Two 20 mg BID Rolling-Six Design 8 weeks between cohorts 10 patients 19 patients Second Stage of Phase if 1 of 10 respond All CMML WHO subtypes were included without regard to previous therapy. Key exclusion criteria included an ANC < 0.25x103 c/dl and a platelet count < 35x103c/dL. Phase 1 completed, n=20 pts (Padron, et al Clinical Cancer Res 2016) Deep Sequencing of recurrent gene mutations in CMML before and after therapy. Comprehensive Cytokine profiling Padron et al for MDS CRC, ASH 2017, Abstract # 162

Summary of Responses Three bi- and tri- lineage hematologic responses by MDS IWG 2006 (i.e. 2 HI-E, 2 HI-P, 1 HI-N) One bone marrow complete response identified One partial marrow response identified 6 of 13 patients (46%) with splenomegaly had a > 50% reduction by physical exam When using clinical benefit as defined by the MDS/MPN- IWG response criteria 11 of 24 (46%) evaluable patients responded *Most common reason for discontinuation was disease progression (n=10)* Padron et al for MDS CRC, ASH 2017, Abstract # 162

Ruxolitinib-treated CMML compared to historical controls Hazard Ratio= 0.42 P<0.005 *When adjusted for age, Global MD Anderson Model, WHO subtype, and mean interval from diagnosis until treatment* Padron et al for MDS CRC, ASH 2017, Abstract # 162 23

Other Agents in CMML Tipifarnib: Farnesyltransferase inhibitor. FTs are necessary for proper Localization of RAS molecules to the inner cell membrane, hypothesized to be more effective in WT RAS patients. 15 patients treated, 11 with CMML-1 and 4 with CMML-2. Commons AEs were thrombocytopenia, diarrhea, nausea Only 1 of 7 patients evaluable for response had marrow response. Patnaik M, ASH abstract 2963, 2017. Eltrombopag: Tested in 25 CMML patients with lower risk features and platelets <50,000. Itzykson R, ASH Abstract 4266, 2017. 63% had HI-Platelets with median duration of response of 8 months.

Azacitidine Survival Study (AZA-001) 5AC 75 mg/m 2 /d x 7 d q28 d (n=179) Screening/Central Pathology Review Investigator CCR Tx Selection Randomization (n=358) Best Supportive Care (BSC) (n=105) Low Dose Ara-C (LDAC, (20 mg/m2/d x 14d q28-42) (n=49) Std Chemo (7 + 3) (n=25) BSC was included with each arm. Tx continued until unacceptable toxicity, AML transformation, or disease progression Fenaux et al. Lancet Oncology 2009;10:223..

Proportion Surviving Overall Survival: Azacitidine vs CCR 1.0 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 24.4 months 15 months 0.1 0.0 0 5 10 15 20 25 30 35 40 Time (months) from Randomization Log-Rank p=0.0001 HR = 0.58 [95% CI: 0.43, 0.77 Difference: 9.4 months 5AC** CCR **Improved OAS also seen for low blast count AML Fenaux et al. Lancet Oncology 2009;10:223..

Patients with MDS for whom hypomethylating agents have failed have poor outcomes overall. Bejar R, and Steensma D P Blood 2014;124:2793-2803 2014 by American Society of Hematology

There remains no standard of care after hypomethylating agent failure: Omecetaxine phase II (Short et al, ASH abstract 2967, 2017). 35 patients, median BM blasts 10% OM given at 1.25 mg/m2 SQ q 12 hours x 3 days on 28 day cycle 12/35 patients responded: 3 CRp and 9 Cri Fatigue and nausea were most common Aes Median OS was 7.6 months

NOVEL APPROACHES IN MDS CC-486: Oral Vidaza, ongoing trials. ASTX727 (oral decitabine) paired with E7727 an oral cytidine deaminase inhibitor. The pairing Assures that oral decitabine will not be degraded, favorable PK have been reported (Garcia- Manero, ASH Abstract 4274, 2017). SGI-110 (guadecitabine): Phase III is ongoing. Rigosertib (Multikinase inhibitor in HMA failure). We are participating in a large phase III trial. Randomized versus investigator choice. LSD1 inhibitors: We are participating in a phase I in AML, run by lung cancer group. IDH1/IDH2 inhibition: These mutations present in 15% of AML but only 6% of MDS. They lead to aberrant hypermethylation. In the phase I/II study in advanced myeloid malignancies 6/16 MDS patients had a response (1 CR, 1 PR, and 4 HI). Stein E, ASH abstract 343, 2016. Venetoclax (BCL2 inhibition): Ongoing clinical trials in up front and HMA failure settings.

Clinical Trials for MDS Open At Penn Up front: Azacitadine plus pracinostat. IV Vidaza (7 day dosing) plus oral pracinostat. HMA failure: Rigosertib 2:1 randomization versus best available therapy. BMT-CTN 1102: Hypomethylating agent/best supportive care versus reduced intensity conditioning allo SCT for MDS ages 50-75 IPSS 1.5 or greater. *Observational only *Biological randomization based on presence or absence of matched sibling or 10/10 unrelated donor.