Presentation is loading. Please wait.

Presentation is loading. Please wait.

Srdan Verstovsek, MD, PhD Professor

Similar presentations


Presentation on theme: "Srdan Verstovsek, MD, PhD Professor"— Presentation transcript:

1 Myelofibrosis and Polycythemia Vera: Current and Emerging Treatment Options
Srdan Verstovsek, MD, PhD Professor Division of Cancer Medicine Department of Leukemia The University of Texas M. D. Anderson Cancer Center Houston, Texas This activity is supported by an educational grant from Incyte.

2 About These Slides Please feel free to use, update, and share some or all of these slides in your noncommercial presentations to colleagues or patients When using our slides, please retain the source attribution: These slides may not be published, posted online, or used in commercial presentations without permission. Please contact for details Slide credit: clinicaloptions.com Disclaimer: The materials published on the Clinical Care Options Web site reflect the views of the authors of the CCO material, not those of Clinical Care Options, LLC, the CME providers, or the companies providing educational grants. The materials may discuss uses and dosages for therapeutic products that have not been approved by the United States Food and Drug Administration. A qualified healthcare professional should be consulted before using any therapeutic product discussed. Readers should verify all information and data before treating patients or using any therapies described in these materials.

3 Faculty Disclosures Srdan Verstovsek, MD, PhD, has disclosed that his institution has received funds for the conduct of clinical studies that he participated in from AstraZeneca, Blueprint Medicines, Bristol-Myers Squibb, Celgene, CTI BioPharma, Galena BioPharma, Genentech, Geron, Gilead Sciences, Incyte, Lilly Oncology, NS Pharma, Pfizer, Promedior, Roche, and Seattle Genetics. This slide lists disclosures for the faculty who was involved in the production of these slides.

4 Polycythemia Vera

5 Case 1: Pt With PV Requiring First-line Cytoreductive Therapy
A 51-yr-old postmenopausal woman with PV Good control of hematocrit; no need for phlebotomy for last 2 yrs Hematocrit: 42%; platelets: 945 x 109/L; WBC: 12 x 109/L; chemistry normal; iron deficiency History of hypertension and depression She complains about significant itching that affects quality of life, no relief from antihistamines or increased dose of aspirin, increasing fatigue On physical exam, you feel spleen 3 cm below left costal margin PV, polycythemia vera; WBC, white blood cell. Slide credit: clinicaloptions.com

6 Case 1: Expert Recommendations
51-yr-old woman with PV; hematocrit controlled; PMH of HTN and depression; complains about significant itching, increasing fatigue HTN, hypertension; PMH, past medical history; PV, polycythemia vera. Available at: clinicaloptions.com/MPNTool

7 Thrombosis Risk-Adapted Management of PV
Category Characteristics Treatment Low risk Age ≤ 60 yrs AND no history of thrombosis Therapeutic phlebotomy (goal Hct < 45%) ASA 81 mg daily Address CV modifiable risk factors High risk Age > 60 yrs OR history of thrombosis All the above, AND Cytoreductive therapy First line Hydroxyurea IFN-α Busulfan* Second line Ruxolitinib CV, cardiovascular; Hct, hematocrit; IFN, interferon; PV, polycythemia vera. *For pts > 70 yrs of age. Indications for cytoreduction in low-risk pts with: Frequent phlebotomy requirement – Platelets > 1500 x 109/L (risk of bleeding) Progressive leukocytosis – Severe disease-related symptoms Slide credit: clinicaloptions.com Barbui T, et al. J Clin Oncol. 2011;29: Tefferi A, et al. Am J Hematol. 2015;90:

8 European LeukemiaNet Clinicohematologic Response in PV
Response Grade Response in PV CR Hematocrit < 45% without phlebotomy and Platelet count ≤ 400 x 109/L and WBC count ≤ 10 x 109/L and Normal spleen size on imaging and No disease-related symptoms PR Hematocrit < 45% without phlebotomy or Response in ≥ 3 of the other criteria No response Any response that does not satisfy PR PV, polycythemia vera; WBC, white blood cell. *Disease-related symptoms include microvascular disturbances, pruritus, and headache. Slide credit: clinicaloptions.com Barbui T, et al. J Clin Oncol. 2011;29:

9 CYTO-PV: Intensity of Treatment and Cardiovascular Events in PV
Multicenter, randomized, open-label phase III trial in 26 Italian centers Primary endpoint: time to major thrombotic events or death from CV causes Low hematocrit target (Hct < 45%) ASA + more intensive phlebotomy and/or cytoreductive drugs* per investigator decision (n = 182) Pts with JAK2-positive PV being treated with phlebotomy, hydroxyurea, or both; no substantially shortened life expectancy (N = 365) High hematocrit target (Hct 45% to 50%) ASA + less intensive phlebotomy and/or cytoreductive drugs* per investigator decision (n = 183) ASA, aspirin; CV, cardiovascular; Hct, hematocrit; PV, polycythemia vera. *Hydroxyurea recommended for pts with progressive thrombocytosis, splenomegaly, or high risk of thrombosis. Slide credit: clinicaloptions.com Marchioli R, et al. N Engl J Med. 2013;368:22-33.

10 CYTO-PV: Total CV Events by Hematocrit Target
1.0 0.8 Low Hct High Hct P = .01 0.6 Probability of Remaining Event Free 0.4 Events, n/N (%) HR (95% CI) Low Hct 8/182 (4.4) High Hct /183 (10.9) ( ) CV, cardiovascular; Hct, hematocrit. 0.2 6 12 18 24 30 36 42 48 Mos Mean follow-up: 28.9 mos Slide credit: clinicaloptions.com Marchioli R, et al. N Engl J Med. 2013;368:22-33.

11 Time to Thrombosis in PV Pts Treated With HU Requiring Ongoing Therapeutic Phlebotomy
Retrospective analysis of observational data from first 5 yrs of hydroxyurea + phlebotomy in pts with PV (N = 533) 100 80 60 Thrombosis (%) 40 HU, hydroxyurea; PV, polycythemia vera. ≥ 3 phlebotomies/yr P = .0001 20 < 3 phlebotomies/yr 12 24 36 48 60 Mos on Therapy Slide credit: clinicaloptions.com Alvarez-Larrán A, et al. Haematologica. 2017;102:

12 Characteristics and Treatment of PV Pts With HU in Clinical Practice
Retrospective chart review at 34 German centers (N = 1476) HU Doses Used Response Rates to HU Reasons for HU Resistance (n = 306) 6.3% Constitutional Symptoms 20% 32.2% Splenomegaly 8% 43.4% 50.3% 67.8% WBC/PLT Elevation 41% HU, hydroxyurea; PLT, platelet; PV, polycythemia vera; WBC, white blood cell. Phlebotomy 77% < 1 g/day 1-2 g/day > 2 g/day Sensitive Resistant 20 40 60 80 100 Pts (%) Slide credit: clinicaloptions.com Jentsch-Ullrich K, et al. J Cancer Res Clin Oncol. 2016;142:

13 PVN1: PegIFN as Initial Cytoreductive Therapy for PV
Multicenter, single-arm phase II trial: pts with PV with no prior treatment or < 2 yrs phlebotomy/cytoreductive therapy treated with ASA QD + pegIFN SC QW (N = 37)[1] 78% still on pegIFN alone after median follow-up of 31 mos 12-mo hematologic CR rate: 95% 12-mo AE rate: 89%; treatment discontinued in 35% (24% for toxicity) AEs mostly grade 1/2, decreased over time JAK2 V617F allele burden CR in 7/29 (24%) of pts treated with pegIFN alone PR in 14/29 (48%) Targets JAK2 V617F clones without affecting TET2-mutant cells[2] 100 6 12 18 24 30 36 80 60 JAK2 V617F Circulating Alleles (%) 40 20 AE, adverse event; ASA, aspirin; pegIFN, peginterferon; PV, polycythemia vera. Mos Slide credit: clinicaloptions.com 1. Kiladjian JJ, et al. Blood. 2008;112: Kiladjian JJ, et al. Leukemia. 2010;24:

14 Planned interim analysis conducted when 75 pts treated for 1 yr
MPD-RC 112: First-line PegIFN vs HU for High-Risk PV and Essential Thrombocythemia Multicenter, randomized, open-label phase III study[1] Primary endpoint: CR by ELN criteria at 12 mos[2] Mo 12 Hydroxyurea 500 mg BID PO (n = 86) Pts with high-risk* PV/ET diagnosed < 5 yrs prior and no prior treatment† (N = 168) Planned interim analysis conducted when 75 pts treated for 1 yr PegIFN α-2a 180 mcg QW SC‡ (n = 82) ET, essential thrombocythemia; ELN, European LeukemiaNet; HU, hydroxyurea; pegIFN, peginterferon; PV, polycythemia vera. *Defined as age ≥ 60 yrs; prior thrombosis, erythromelalgia, or migraine (or hemorrhage for ET); splenomegaly > 5 cm or symptomatic; platelets > 1000 x 109/L for PV or > 1500 x 109/L for ET; or diabetes or hypertension requiring treatment. †Hydroxyurea for < 3 mos permitted. ‡Starting dose of 45 mcg QW gradually increased to maximum dose of 180 mcg QW. Slide credit: clinicaloptions.com 1. Mascarenhas JO, et al. ASH Abstract Barosi G, et al. Blood. 2009;113:

15 MPD-RC 112: Key Findings Mo 12 Response Hydroxyurea (n = 39)
PegIFN α-2a (n = 36) P Value* PR CR ORR All pts, n (%) 14 (36) 13 (33) 27 (69) 19 (53) 10 (28) 29 (81) .6 ET subset, n/N (%) 4/16 (25) 7/16 (44) 11/16 (69) 6/15 (40) 12/15 (80) .8 PV subset, n/N (%) 10/23 (44) 6/23 (26) 16/23 (70) 13/21 (62) 4/21 (19) 17/21 (81) AE,† n (%) Hydroxyurea (n = 36) PegIFN α-2a (n = 36) P Value Any AE 32 (89) 36 (100) .04 AE grade ≥ 3 5 (14) 17 (47) .002 Depression 10 (28) < .001 Dyspnea 1 (3) 7 (19) .02 Fatigue 18 (50) .05 Flulike symptoms 12 (33) Injection-site reaction 9 (25) .001 Pruritus 3 (8) .03 AE, adverse event; ET, essential thrombocythemia; pegIFN, peginterferon; PV, polycythemia vera. *P value for comparison of CR across arms. †Showing AEs that occurred in ≥ 15% of pts in either arm and for which there was a significant difference in rates between study arms. Slide credit: clinicaloptions.com Mascarenhas JO, et al. ASH Abstract 479.

16 Use of PegIFN to Treat PV: Expert Recommendations
Pt with high-risk PV for whom first-line cytoreductive therapy is required; no factors that would preclude pegIFN use PegIFN, peginterferon; PV, polycythemia vera Available at: clinicaloptions.com/MPNTool

17 Case 2: Pt With Inadequate Response to HU
68-yr-old man diagnosed with PV 3 yrs ago Characteristic Finding Past treatments Has been managed successfully with low-dose ASA, phlebotomy, and HU (500 mg BID; higher dose caused mouth ulcers) Past medical history Diabetes, depression Current visit Presents with complaints of fatigue, pruritus, and occasional night sweats Hb: 13.7 g/dL Hct: 43% WBC count: 16.4 x 109/L Platelets: 640 x 109/L Peripheral smear: leukocytosis, thrombocytosis, no nucleated RBCs, no blasts Ferritin decreased, TIBC increased, MCV low; chemistry WNL Physical exam reveals splenomegaly of 5 cm below left costal margin Bone marrow biopsy: 1+ out of 3+ fibrosis, normal cytogenetics ASA, aspirin; Hb, hemoglobin; Hct, hematocrit; HU, hydroxyurea; MCV, mean corpuscular volume; PV, polycythemia vera; RBC, red blood cell; TIBC, total iron binding capacity; WBC, white blood cell; WNL, within normal limits. Slide credit: clinicaloptions.com

18 Case 2: Expert Recommendations
68-yr-old man with fatigue, pruritus, and night sweats despite hydroxyurea therapy; splenomegaly of 5 cm below left costal margin; history of depression Available at: clinicaloptions.com/MPNTool

19 ELN Criteria for Hydroxyurea Resistance and Intolerance in PV
Need for phlebotomy (Hct <45%) Platelets > 400 x 109/L and WBC > 10 x 109/L No reduction of spleen by 50% No reduction of spleen symptoms After > 3 mos at MTD or 2 g/day Cytopenias (any) ANC < 1.0 x 109/L Hemoglobin x 109/L Platelets < 100 x 109/L At lowest dose to achieve either a PR or CR Intolerance/Resistance ANC, absolute neutrophil count; ELN, European LeukemiaNet; GI, gastrointestinal; Hct, hematocrit; MTD, maximum tolerated dose; PV, polycythemia vera; WBC, white blood cell. GI toxicity Fevers Mucocutaneous toxicity Skin cancers At any dose Barosi G et al. Br J Haematol. 2010;148:

20 Mean MPN-10 Symptom Scores by Known HU Use
Symptom Profiles of PV Pts by HU Use: Implications for Inadequately Controlled Disease Prospective analysis of symptom burden in pts with PV (N = 1334) Mean MPN-10 Symptom Scores by Known HU Use Abdominal discomfort * Bone pain * Known HU use No known HU use Concentration * Early satiety * Fatigue * Symptom Fever HU, hydroxyurea; MPN-10, Myeloproliferative Neoplasm Symptom Assessment Form; PV, polycythemia vera. Inactivity * Itching *P < .001 †P < .05 Night sweats * Weight Loss 1 2 3 4 5 6 7 8 9 10 Slide credit: clinicaloptions.com Geyer H, et al. J Clin Oncol. 2016;34:

21 Time to Transformation to MF or AML in PV Pts Treated With Hydroxyurea
Retrospective analysis of time to myelofibrotic transformation and cytopenia for median follow-up of 4.6 yrs in pts with PV (N = 890) Myelofibrosis Acute Myeloid Leukemia 100 100 HU intolerance/resistance No HU intolerance/resistance Cytopenia No cytopenia 80 80 60 60 Pts (%) Pts (%) P = .03 40 40 AML, acute myeloid leukemia; HU, hydroxyurea; MF, myelofibrosis; PV, polycythemia vera. 20 20 P < .001 5 10 15 20 25 30 5 10 15 20 25 30 Yrs Yrs Slide credit: clinicaloptions.com Alvarez-Larrán A, et al. Br J Haematol. 2016;172:

22 Recommendations for Second-line PV Therapy
Pt Characteristics Options Inadequate response or intolerance to HU Ruxolitinib, IFN-α[1,2] Inadequate response or intolerance to IFN-α HU[1] Short life expectancy Busulfan, pipobroman, or 32P[1] HU, hydroxyurea; IFN, interferon; PV, polycythemia vera. 1. Barbui T, et al. J Clin Oncol. 2011;29: 2. Ruxolitinib [package insert]. Slide credit: clinicaloptions.com

23 PegIFN for Pts With Advanced PV
Open-label, single-arm phase II trial in which pts with PV treated with pegIFN (n = 43) Baseline characteristics: median age, 54 yrs; PV duration, 50 mos; previous cytoreductive therapy, 49%; median prior therapies, 1 (range: 0-4) Median follow-up, entire cohort*: 42 mos 100 80 Outcome Pts (n = 43) Overall hematologic response, % 79 Complete hematologic response, % 76 Median time to CR, days (range) 40 (3-1478) Complete molecular response,† % 18 Partial molecular response,‡ % 35 Drug-related tx discontinuation, % 20 64 60 53 JAK2 V617F Circulating Alleles (%) 41 40 ET, essential thrombocythemia; pegIFN, peginterferon; PV, polycythemia vera; tx, treatment. 29 20 19 16 17 13 9 8 4.5 *Included an additional 40 pts with ET. †Undetectable JAK2V617F allele burden. ‡> 50% decrease. 6 12 18 24 30 36 42 48 54 60 Mos Slide credit: clinicaloptions.com Quintás-Cardama A, et al. Blood. 2013;122:

24 PegIFN for Pts With Advanced PV: Long-term Follow-up
Post hoc analysis of open-label, single-arm phase II trial in which pts with PV treated with pegIFN (n = 43) Median follow-up: 83 mos Response Median hematologic response duration: 65 mos Median molecular response duration: 58 mos Therapy failures occur (vascular events and progression) Safety* Toxicity continued over time (new grade 3/4 events in 10% to 17% of pts/yr) Discontinuations in overall study population: 61% (22% for overt toxicity) PegIFN, peginterferon; PV, polycythemia vera. Slide credit: clinicaloptions.com Masarova L, et al. Lancet Haematol. 2017;4:e165-e175.

25 RESPONSE: Ruxolitinib vs Standard Therapy in Pts With HU Resistance/Intolerance
Ruxolitinib: Janus kinase (JAK) 1 and 2 inhibitor International, multicenter, randomized, open-label phase III study Primary endpoint: proportion with Hct control + spleen volume reduction ≥ 35% (Wk 32) Stratified by HU status (resistance vs intolerance) Wk 32 Ruxolitinib Initial 10 mg BID; dose titrated to maintain Hct (n = 110) Pts with PV requiring phlebotomy; HU resistance/intolerance; spleen volume ≥ 450 cm3, no JAK inhibitor experience, Hct 40% to 45% before randomization* (N = 222) Primary analysis data cutoff at Wk 48 or treatment discontinuation ASA, aspirin; Hct, hematocrit; HU, hydroxyurea; IFN, interferon; PD, progressive disease; PV, polycythemia vera. Crossover at PD or Wk 32 if primary endpoint not met Standard Therapy selected by investigator† (n = 112) All pts received low-dose ASA. *Pts with Hct < 40% or > 50% entered Hct control period prior to randomization. †Excluding 32P, busulfan, and chlorambucil. Slide credit: clinicaloptions.com Vannucchi AM, et al. N Engl J Med. 2015;372:

26 RESPONSE: Key Efficacy Findings at Wk 32
Study design adopted form Rux figures 70 Ruxolitinib Standard therapy 60.0 60 P < .001 Odds ratio: 28.6 (95% CI: ) 50 40 38.2 Pts (%) 30 20.9 19.6 20 10 0.9 0.9 Composite Primary Endpoint ≥ 35% Reduction in Spleen Volume Hematocrit Control Complete hematologic response also significantly improved with ruxolitinib vs standard therapy (23.6% vs 8.9%; P = .003) Slide credit: clinicaloptions.com Vannucchi AM, et al. N Engl J Med. 2015;372:

27 Mean Change From Baseline at Wk 32 GHS/QoL and Functional Subscales
RESPONSE: Mean Change From Baseline in QoL and Disease‐Related Symptoms at Wk 32 Ruxolitinib* Standard therapy* 15 Improvement 10 5 Mean Change From Baseline at Wk 32 Improvement -5 -10 -15 Social Role Pain GHS, global health status; QoL, quality of life. Fatigue GHS/QoL Physical Emotional Cognitive Insomnia Dyspnea Diarrhea Appetite loss Financial difficulties Constipation Nausea and vomiting GHS/QoL and Functional Subscales Individual Symptoms *Pts with available data at both time points: ruxolitinib, n = 86-90; standard therapy, n = Slide credit: clinicaloptions.com Mesa R, et al. Eur J Haematol. 2016;97:

28 RESPONSE: Durability of Hematocrit Control With Ruxolitinib
Graph adopted from RUX RUX3011_figs2ab-3 Preplanned analysis after all pts discontinued or completed Wk 80 study visit 1.0 0.8 0.6 Probability of Remaining Event Free Probability of maintaining Hct control in ruxolitinib arm for ≥ 80 wks from time of response was 0.89 0.4 Hct, hematocrit. 0.2 6 12 18 24 30 36 42 48 54 60 66 72 78 84 90 96 102 108 114 120 126 132 138 144 Wks Pts at risk, n Events, n 25 25 25 25 25 25 24 1 22 2 21 2 20 2 20 2 16 2 16 2 152 152 152 142 102 102 7 2 6 2 5 2 1 2 1 2 2 Slide credit: clinicaloptions.com Verstovsek S, et al. Haematologica. 2016;101:

29 RESPONSE: Thromboembolic AEs at Wk 80
Longer exposure in ruxolitinib arm vs standard therapy arm (227.7 vs 73.6 pt-yrs) Event Rate per 100 Pt-Yrs of Exposure Ruxolitinib (n = 110) Standard Therapy (n = 111*) All Grades Grade 3/4 All thromboembolic events 1.8 0.9 8.2 2.7 Portal vein thrombosis 0.4 Cerebral infarction Ischemic stroke Retinal vascular thrombosis Myocardial infarction 1.4 Deep vein thrombosis Pulmonary embolism Splenic infarction Thrombophlebitis Thrombosis AE, adverse event. *1 pt did not receive study treatment. Slide credit: clinicaloptions.com Verstovsek S, et al. Haematologica. 2016;101:

30 RESPONSE: Ruxolitinib Dose at Wk 32
Starting dose was 10 mg BID At Wk 32, 55 of 98 assessed pts (56%) in ruxolitinib arm were receiving doses of mg BID Most dose adjustments occurred within first 8 wks of treatment Dose adjustments based on CBC monitoring, particularly Hct, white cells, and platelets CBC, complete blood count; Hct, hematocrit. < 10 mg BID 10 mg BID 15 mg BID 20 mg BID 25 mg BID Dose at Wk 32* *Maximum dose within 2-week window of Wk 32 study visit. Slide credit: clinicaloptions.com Vannucchi AM, et al. N Engl J Med. 2015;372:

31 RESPONSE-2: Ruxolitinib vs Best Available Therapy in Pts Without Splenomegaly
International, multicenter, randomized, open-label phase IIIb study in which pts with HU-resistant/intolerant PV who required phlebotomy and had no splenomegaly were treated with ruxolitinib or best available therapy (N = 149) Outcome, Wk 28 Ruxolitinib (n = 74) BAT (n = 75) P Value Hct control,* n (%) 46 (62) 14 (19) < .0001 Complete hematologic response, n (%) 17 (23) 4 (5) .0019 Complete resolution in symptoms, n/N† (%) 17/34 (50) 2/26 (8) NR ≥ 50% reduction in MPN-SAF TSS, n/N (%) 29/64 (45) 5/22 (23) BAT, best available therapy; Hct, hematocrit; HU, hydroxyurea; MPN-SAF TSS, Myeloproliferative Neoplasm Symptom Assessment Form Total Symptom Score; NR, not reported; PV, polycythemia vera. *Primary endpoint. †Pts with baseline MPN-SAF TSS of ≥ 20. Slide credit: clinicaloptions.com Passamonti F, et al. Lancet Oncol. 2017;18:88-99. 31

32 Myelofibrosis

33 Case 3: Pt With MF Who Is a Potential Transplantation Candidate
A 58-yr-old man living with JAK2 V617F-negative primary MF for a number of yrs Characteristic Finding Past treatments Erythropoietin, androgens, HU, thalidomide plus prednisone Anemic, transfused PRBC every 4 mos Current laboratory values Anemic Platelets: 126 x 109/L WBC 17 x 109/L 2% blood blasts Physical examination and interview ECOG PS: 2 Has lost 15 kg in past yr and is very fatigued Experiencing night sweats, low grade fever, bone aches Splenomegaly of 15 cm below costal margin ECOG, Eastern Cooperative Oncology Group; HU, hydroxyurea; MF, myelofibrosis; PRBC, packed red blood cell; PS, performance status; WBC, white blood cell. Slide credit: clinicaloptions.com

34 MF Treatment Is Based on Risk and MF-Related Symptoms/Signs
Minimally symptomatic observation or interferon Low Risk Many symptoms ruxolitinib Intermediate-1 Risk Ruxolitinib or anemia treatment and/or allogeneic HSCT Intermediate-2 Risk Allogeneic HSCT or ruxolitinib and/or anemia treatment HSCT, hematopoietic stem cell transplantation; MF, myelofibrosis. High Risk Allogeneic HSCT or ruxolitinib and/or anemia treatment Mesa RA. Leuk Lymphoma. 2013;54: Geyer HL, et al. Hematology Am Soc Hematol Educ Program. 2014;2014: Slide credit: clinicaloptions.com

35 International Prognostic Scoring System: Risk Classification of MF at Presentation
Risk factors Older than 65 yrs of age Constitutional symptoms Hb < 10 g/dL WBC count > 25 x 109/L Peripheral blood blasts ≥ 1% No. Risk Factors Risk Group Median Survival, Yrs Low 11.3 1 Intermediate-1 7.9 2 Intermediate-2 4.0 ≥ 3 High 2.3 Hb, hemoglobin; MF, myelofibrosis, WBC, white blood cell. Slide credit: clinicaloptions.com Cervantes F, et al. Blood. 2009;113:

36 Prognostic Impact of Driver and High Molecular Risk Nondriver Mutations in Primary MF
Analysis of association between driver mutations and survival in pts with primary MF (N = 617)[1] Analysis of association between set of nondriver mutations (IDH, EZH2, ASXL1, SRSF2) and survival in pts with PMF (N = 797)[2] Presence of mutations predicted decreased survival; ≥ 2 mutations predicted worst survival Driver Mutation Pts, % Median OS, Yrs CALR mutated 22.7 17.7 JAK2 mutated 64.7 9.2 MPL mutated 4.0 9.1 Triple negative 8.6 3.2 MF, myelofibrosis. Slide credit: clinicaloptions.com 1. Rumi E, et al. Blood. 2014;124: Guglielmelli P, et al. Leukemia. 2014;28:

37 Allogeneic HSCT: Why We Prognosticate
Consider HSCT in younger, higher- risk pts whose survival is expected to be < 5 yrs Very few MF pts undergo HSCT Traditionally limited to pts younger than 60 yrs of age and those with HLA-identical sibling match; now possible up to age 75 High transplant-related mortality and morbidity associated with transplantation due to acute and chronic graft vs host disease[2] 1-yr nonrelapse mortality rate: 12% (completely matched donors) to 38% (mismatched) 5-yr survival rate: 56% (matched sibling donors) to 34% (partially matched/mismatched) Risk Group[1] Median Survival, Yrs Low 11.3 Intermediate-1 7.9 Intermediate-2 4.0 High 2.3 HSCT, hematopoietic stem cell transplantation; MF, myelofibrosis. 1. Cervantes F, et al. Blood. 2009;113: 2. Kröger NM, et al. Leukemia. 2015;29: Slide credit: clinicaloptions.com

38 Case 4: Transplant-Ineligible Pt With High-Risk MF
You are taking over care from a retiring doctor of a 76-yr-old man with MPL mutation–positive primary MF Transplantation is not being considered Characteristic Finding Past treatments Hydroxyurea, prednisone, danazol, and thalidomide Current visit Significant constitutional symptoms and weight loss Hb: 8.6 g/dL without transfusions WBC count: 19 x 109/L Platelets: 189 x 109/L EPO level: 125 mU/mL 2% blood blasts Liver is enlarged 4 cm and spleen 9 cm by palpation EPO, erythropoietin; Hb, hemoglobin; MF, myelofibrosis; WBC, white blood cell. Slide credit: clinicaloptions.com

39 Case 4: Expert Recommendations
76-yr-old man with several previous treatments, anemia, 2% blood blasts, EPO mU/mL, platelets 189 x 109/L, splenomegaly, and MF symptoms EPO, erythropoietin; MF, myelofibrosis. Available at: clinicaloptions.com/MPNTool

40 Main Clinical Complications in MF
Pts (%) Anemia (Hb < 10 g/dL) 36 Leukocytosis (> 25 x 109/L) 10 16 Thrombocytopenia (< 100 x 109/L) Splenomegaly 83 Hepatomegaly 65 Thrombosis 7 Hb, hemoglobin; MF, myelofibrosis. Constitutional symptoms 27 Leukemia transformation 13 Passamonti F, et al. Blood. 2010;115: Barbui T, et al. Blood. 2010;115: Passamonti F, et al. Blood. 2010;116: Slide credit: clinicaloptions.com

41 Symptomatic Burden in MF
Mean MPN-SAF Results From Surveys of Pts With MF (N = 96) Symptoms related to: Insomnia 74 Role/ functioning Inactivity 77 Fatigue 99 Itching 54 Myeloproliferation Bone pain 55 Symptom Cough 55 Abd discomfort 72 Splenomegaly Early satiety 76 Abd, abdominal; MF, myelofibrosis; MPN-SAF, Myeloproliferative Neoplasm Symptom Assessment Form. Fever 29 Constitutional symptoms Weight loss 49 Night sweats 63 10 20 30 40 50 60 70 80 90 100 Pts (%) Slide credit: clinicaloptions.com Scherber R, et al. Blood. 2011;118:

42 Needs-Oriented Therapy for MF
Clinical Issue Treatments Anemia ESAs Corticosteroids Danazol Thalidomide, lenalidomide (IMIDs) Symptomatic splenomegaly Ruxolitinib Hydroxyurea Cladribine, IMIDs Splenectomy Constitutional symptoms/QoL Extramedullary hematopoiesis Radiation therapy Hyperproliferative (early) disease Interferon Risk of thrombosis Low-dose aspirin Accelerated/blastic phase Hypomethylating agents Improved survival Allogeneic HSCT ASA, acetylsalicylic acid; ESA, erythropoiesis-stimulating agents; HSCT, hematopoietic stem cell transplantation; IMID, immunomodulatory drug; MF, myelofibrosis. Slide credit: clinicaloptions.com

43 COMFORT-I and -II: Ruxolitinib for Pts With Intermediate-2–Risk/High-Risk MF
Randomized phase III studies in which pts with intermediate-2–risk/high-risk MF were treated with ruxolitinib (15 or 20 mg BID) vs placebo (COMFORT-I, N = 309) or best available therapy (COMFORT-II, N = 149) Outcome COMFORT-I, Wk 24[1] P Value COMFORT-II, Wk 48[2] Ruxolitinib (n = 155) Placebo (n = 154) Ruxolitinib (n = 144) BAT (n = 73) Spleen volume reduction ≥ 35%,* % 41.9 0.7 < .001 28 ≥ 50% reduction in MF-SAF TSS, % 45.9 5.3 NR D/c for AEs 11.0 10.6 8 5 AE, adverse event; BAT, best available therapy; D/c, discontinued; MF, myelofibrosis; NR, not reported. *Primary endpoint. †n = 151. Grade 3/4 anemia/thrombocytopenia/neutropenia in COMFORT-I, % Ruxolitinib, 45/13/7 ; placebo 19/1/2† 1. Verstovsek S, et al. N Engl J Med. 2012;366: 2. Harrison C, et al. N Engl J Med. 2012;366: Slide credit: clinicaloptions.com 43

44 COMFORT-I: 5-Yr OS With Ruxolitinib vs Placebo
Deaths, n/N (%) Censored, n/N (%) Median OS, Wks Median F/U, Wks RUX PBO 69/155 (44.5) 82/154 (53.2) 86/155 (55.5) 72/154 (46.8) NR 200 268.4 269.0 1.00 0.75 Probability of OS 0.50 Median time to crossover from PBO to RUX: 39.9 wks 0.25 F/U, follow-up; NR, not reached; PBO, placebo; RUX, ruxolitinib. HR: 0.69 (95% CI: ; P = .025 [nominal]) 24 48 72 96 120 144 168 192 216 240 264 Wks Pts at Risk, n RUX PBO 155 154 148 144 137 119 124 105 112 95 108 85 100 78 86 72 80 59 75 51 69 46 57 38 Slide credit: clinicaloptions.com Verstovsek S, et al. J Hematol Oncol. 2017;10:55.

45 COMFORT-II: 5-Yr OS With Ruxolitinib vs BAT
Median OS, Yrs 1.0 RUX NR BAT (ITT) BAT (RPSFT*) 2.7 0.8 0.6 Probability of OS 0.4 0.2 HR: 0.67 (95% CI: ; P = .06) BAT, best available therapy; ITT, intent to treat; NR, not reached; RPSFT, rank-preserving structural failure time; RUX, ruxolitinib. 1 2 3 4 5 6 Pts at Risk, n RUX BAT (ITT) BAT (RPSFT*) Yrs 0 0 0 *PRSFT modeling estimates treatment effect corrected for crossover. Median follow-up: 4.3 yrs; majority crossover from best available therapy to ruxolitinib Slide credit: clinicaloptions.com Harrison CN, et al. Leukemia. 2016;30:

46 COMFORT-I and -II Exploratory Analysis: Impact of Anemia on OS
Anemia at BL associated with decreased OS in both arms New or worsening anemia (after initiating ruxolitinib) did not affect OS (HR: 1.030) OS improved with ruxolitinib vs control regardless of anemia status at BL HR: (95% CI: ; P = .0102) 1.0 0.8 0.6 0.4 0.2 12 23 35 48 60 72 84 96 108 120 132 144 156 Wks 168 Ctl: anemic at BL Ctl: not anemic at BL Control Probability of OS RUX: anemic at BL RUX: not anemic at BL Ruxolitinib 1.0 0.8 0.6 0.4 0.2 12 23 35 48 60 72 84 96 108 120 132 144 156 Wks 168 Probability of OS No Anemia at BL 1.0 0.8 0.6 0.4 0.2 12 23 35 48 60 72 84 96 108 120 132 144 156 RUX: not anemic at BL Ctl: not anemic at BL Wks 168 Anemia at BL 1.0 0.8 0.6 0.4 0.2 12 23 35 48 60 72 84 96 108 120 132 144 156 Wks 168 RUX: anemic at BL Ctl: anemic at BL BL, baseline; Ctl, control; RUX, ruxolitinib. After Wk 144, curve interpretation affected by low pt n. Slide credit: clinicaloptions.com Gupta V, et al. Haematologica. 2016;101:e482-e484.

47 OS by Degree of Spleen Length Reduction in Pts Receiving Ruxolitinib
Open-label, single-arm phase I/II study (N = 107) 1.0 0.8 0.6 Probability of OS 0.4 < 25% spleen length reduction (n = 23) ≥ 25% but < 50% spleen length reduction (n = 13) ≥ 50% spleen length reduction (n = 61) 0.2 For < 25% vs ≥ 50% spleen length reduction: HR: 0.22 (95% CI: ; P = .0001) 6 12 18 24 30 36 42 48 Slide credit: clinicaloptions.com Verstovsek S, et al. Blood. 2012;120: Mos

48 Tips for Using Ruxolitinib to Treat Pts With MF
Starting dose selected based on platelet number; anemia is NOT contraindication for use Development of anemia DOES NOT affect benefits Decision to stop ruxolitinib will depend on a combination of different factors, including benefit and presence/absence of toxicity Avoid abrupt interruption of ruxolitinib in pts responding well to therapy MF, myelofibrosis. Mesa RA, et al. Int J Hematol. 2016;104: Ruxolitinib [package insert]. Slide credit: clinicaloptions.com

49 Initial Approach to the Treatment of Anemia in Myelofibrosis
EPO levels ADEQUATE ≥ 500 mU/mL INADEQUATE < 500 mU/mL ESA x 3 mos EPO, erythropoietin; ESA, erythropoiesis-stimulating agent. Danazol, others No response Response Mesa RA. Leuk Lymphoma. 2013;54: Cervantes F, et al. Br J Haematol ;127: Kröger N, et al. Leukemia. 2008;22: Slide credit: clinicaloptions.com

50 Splenectomy and Splenic Irradiation for MF
Treatment Indications Contraindications Risks Splenectomy Symptomatic splenomegaly unresponsive to treatment Severe refractory anemia and thrombocytopenia Unresponsive constitutional symptoms Uncontrollable hemolysis Portal hypertension Thrombocytosis Up to 31% operative morbidity Up to 9% perioperative mortality Liver enlargement and failure Higher acute transformation rate Median survival post splenectomy: 27 mos Splenic irradiation Symptomatic splenomegaly in poor candidates for surgery Severe pain from splenic infarction As preparation for splenectomy Long-lasting cytopenias (44%) MF, myelofibrosis. Tefferi A, et al. Blood. 2000;95: Li Z, et al. Leukemia. 2001;15: Elliott MA, et al. Blood Rev. 1999;13: Slide credit: clinicaloptions.com

51 JAK Inhibitors Under Investigation for MF
Agent MoA Key Studies Pacritinib JAK2 and FLT3 kinase inhibitor Phase III PERSIST-1[1,2]: no JAK2 inhibitors in BAT Spleen volume reduction ≥ 35% at Wk 24: pacritinib 19% vs BAT 5% (P = .0003) Improved responses durable out to Wk 72 without cumulative toxicity Phase III PERSIST-2[3]: platelets < 100 x 109/L; JAK2 inhibitors allowed in BAT Spleen volume reduction ≥ 35% at Wk 24: pacritinib (BID or QD) 18% vs BAT 3% (P = .0001) ≥ 50% reduction in MF-SAF TSS at Wk 24: pacritinib (BID or QD) 25% vs BAT 14% (P = .079) No significant differences in OS between groups Clinical trials placed on hold in December 2016 due to concerns about excess pt deaths related to intracranial hemorrhage, cardiac failure, and cardiac arrest; hold has since been removed NS-018 JAK2 inhibitor Ongoing phase I/II study: phase II focus on pts with MPN previously treated with other JAK2 inhibitors[4] BAT, best available therapy; MF, myelofibrosis; MoA, mechanism of action; MPN, myeloproliferative neoplasm. 1. Mesa RA, et al. Lancet Haematol. 2017;[Epub ahead of print]. 2. Mesa RA, et al. ASCO Abstract Mascarenhas J, et al. ASH Abstract LBA ClinicalTrials.gov. NCT Slide credit: clinicaloptions.com

52 Other Agents Under Investigation for MF
MoA Key Studies Imetelstat Telomerase inhibitor Phase II study[1] Imetelstat for JAK inhibitor-treated pts with int-2–risk to high-risk MF suspended due to lack of response LCL 161 SMAC mimetic/inhibitor of apoptosis antagonists Ongoing phase II study[2,3] In preplanned efficacy analysis, 6 of 16 (38%) evaluable pts with MPNs showed objective responses PRM-151 Recombinant PTX-2; antifibrotic Phase II study[4,5] Reductions in bone marrow fibrosis observed in 11 of 26 (43%) evaluable pts at Wk 24; sustained reductions in responders through Wk 72 Many pts had reductions in symptoms and spleen size, improvements in anemia at Wk 24 MF, myelofibrosis; MoA, mechanism of action; MPN, myeloproliferative neoplasm; PBO, placebo; RBC, red blood cell; SMAC, second mitochondria-derived activator of caspases. 1. ClinicalTrials.gov. NCT ClinicalTrials.gov. NCT Pemmaraju N, et al. ASH Abstract Verstovsek S, et al. 2014;124: Verstovsek S, et al. Blood. 2015;126:56. Slide credit: clinicaloptions.com

53 Summary Treatment for MF and PV should be individualized
PV: assess for thrombotic risk; low-risk pts: aim to control Hct < 45% and eliminate disease-related symptoms; high-risk pts: institute cytoreductive therapy and aim to control Hct, WBCs, platelets, splenomegaly and disease-related symptoms First-line cytoreductive treatment: HU, pegIFN For pts with HU resistance/intolerance: ruxolitinib, pegIFN MF: assess potential for transplant; aim to control clinically relevant symptoms and signs Allogeneic HSCT considered for pts with intermediate-2–risk or high-risk MF and life expectancy < 5 yrs Manage splenomegaly and disease-related symptoms (ruxolitinib), and anemia (ESA, danazol) ESA, erythropoiesis-stimulating agents; Hct, hematocrit; HSCT, hematopoietic stem cell transplantation; HU, hydroxyurea; MF, myelofibrosis; pegIFN, peginterferon; PV, polycythemia vera; WBC, white blood cell. Slide credit: clinicaloptions.com

54 Go Online for More CCO Coverage of MF and PV!
CME-certified online text module on the treatment of MF and PV to accompany these slides with expert faculty commentary on all key studies Interactive Treatment Decision Support Tool for MF and PV— see treatment choices of 5 experts for your patient scenarios clinicaloptions.com/oncology


Download ppt "Srdan Verstovsek, MD, PhD Professor"

Similar presentations


Ads by Google