The MDM2 antagonist idasanutlin in patients with polycythemia vera: results from a single-arm phase 2 study

John Mascarenhas, Francesco Passamonti, Kate Burbury, Tarec Christoffer El-Galaly, Aaron Gerds, Vikas Gupta, Brian Higgins, Kathrin Wonde, Candice Jamois, Bruno Kovic, Ling-Yuh Huw, Sudhakar Katakam, Margherita Maffioli, Ruben Mesa, Jeanne Palmer, Marta Bellini, David M Ross, Alessandro M Vannucchi, Abdulraheem Yacoub, John Mascarenhas, Francesco Passamonti, Kate Burbury, Tarec Christoffer El-Galaly, Aaron Gerds, Vikas Gupta, Brian Higgins, Kathrin Wonde, Candice Jamois, Bruno Kovic, Ling-Yuh Huw, Sudhakar Katakam, Margherita Maffioli, Ruben Mesa, Jeanne Palmer, Marta Bellini, David M Ross, Alessandro M Vannucchi, Abdulraheem Yacoub

Abstract

Idasanutlin, an MDM2 antagonist, showed clinical activity and a rapid reduction in JAK2 V617F allele burden in patients with polycythemia vera (PV) in a phase 1 study. This open-label phase 2 study evaluated idasanutlin in patients with hydroxyurea (HU)-resistant/-intolerant PV, per the European LeukemiaNet criteria, and phlebotomy dependence; prior ruxolitinib exposure was permitted. Idasanutlin was administered once daily on days 1 through 5 of each 28-day cycle. The primary end point was composite response (hematocrit control and spleen volume reduction > 35%) in patients with splenomegaly and hematocrit control in patients without splenomegaly at week 32. Key secondary end points included safety, complete hematologic response (CHR), patient-reported outcomes, and molecular responses. All patients (n = 27) received idasanutlin; 16 had response assessment (week 32). Among responders with baseline splenomegaly (n = 13), 9 (69%) attained any spleen volume reduction, and 1 achieved composite response. Nine patients (56%) achieved hematocrit control, and 8 patients (50%) achieved CHR. Overall, 43% of evaluable patients (6/14) showed a ≥50% reduction in the Myeloproliferative Neoplasm Symptom Assessment Form Total Symptom Score (week 32). Nausea (93%), diarrhea (78%), and vomiting (41%) were the most common adverse events, with grade ≥ 3 nausea or vomiting experienced by 3 patients (11%) and 1 patient (4%), respectively. Reduced JAK2 V617F allele burden occurred early (after 3 cycles), with a median reduction of 76%, and was associated with achieving CHR and hematocrit control. Overall, the idasanutlin dosing regimen showed clinical activity and rapidly reduced JAK2 allele burden in patients with HU-resistant/- intolerant PV but was associated with low-grade gastrointestinal toxicity, leading to poor long-term tolerability. This trial was registered at www.clinincaltrials.gov as #NCT03287245.

© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Clinical response in evaluable patients at C3D28, C5D28, week 32, and C11 day 28. (A) Patients with hematocrit (Hct) control. (B) Patients who showed response according to the ELN hematologic response criteria. (C) Patients with CHR (C) or composite response (D). CR, complete remission; PR, partial remission.
Figure 2.
Figure 2.
Change in spleen volume by ruxolitinib exposure. (A) Percentage change in spleen volume from baseline at any time in evaluable patients. (B) Percentage change in spleen volume at week 32 in evaluable patients. (C) Percentage change in spleen volume at C3D28, C5D28, and week 32 in each patient evaluable at the assessment points. SVR, spleen volume reduction.
Figure 3.
Figure 3.
Patient-reported outcomes per MPN-SAF TSS in evaluable patients. (A) Median change from baseline in MPN-SAF TSS at key assessment time points. (B) Median percentage change from baseline in patient-reported scores at C2D1, C3D28, C5D28, and week 32. Error bars represent IQR. (C) Patients with a ≥50% reduction from baseline MPN-SAF TSS. C2D1, C2 day 1; C11D28, C11 day 28; C12D28, C12 day 28; C14D28, C14 day 28; C17D28, C17 day 28; C20D28, C20 day 28.
Figure 4.
Figure 4.
JAK2 allele burden in evaluable patients. (A) JAK2 allele burden at baseline in patients with or without splenomegaly. (B) Percentage change from baseline in JAK2 allele burden in patients with (responders) or without CHR (nonresponders) at C3D28, C5D28, and week 32. (C) Comparison of reduction in JAK2 allele burden between patients with hematocrit control (responders) or without hematocrit control (nonresponders). (D) Comparison of reduction in JAK2 allele burden between patients with CHR (responders) or without CHR (nonresponders). Black stars represent the mean.
Figure 4.
Figure 4.
JAK2 allele burden in evaluable patients. (A) JAK2 allele burden at baseline in patients with or without splenomegaly. (B) Percentage change from baseline in JAK2 allele burden in patients with (responders) or without CHR (nonresponders) at C3D28, C5D28, and week 32. (C) Comparison of reduction in JAK2 allele burden between patients with hematocrit control (responders) or without hematocrit control (nonresponders). (D) Comparison of reduction in JAK2 allele burden between patients with CHR (responders) or without CHR (nonresponders). Black stars represent the mean.

References

    1. Griesshammer M, Sadjadian P. The BCR-ABL1-negative myeloproliferative neoplasms: a review of JAK inhibitors in the therapeutic armamentarium. Expert Opin Pharmacother. 2017;18(18):1929-1938.
    1. Pardanani A, Lasho TL, Finke C, Hanson CA, Tefferi A. Prevalence and clinicopathologic correlates of JAK2 exon 12 mutations in JAK2V617F-negative polycythemia vera. Leukemia. 2007;21(9):1960-1963.
    1. Tefferi A, Barbui T. Polycythemia vera and essential thrombocythemia: 2021 update on diagnosis, risk-stratification and management. Am J Hematol. 2020;95(12):1599-1613.
    1. Vannucchi AM, Antonioli E, Guglielmelli P, Pardanani A, Tefferi A. Clinical correlates of JAK2V617F presence or allele burden in myeloproliferative neoplasms: a critical reappraisal. Leukemia. 2008;22(7):1299-1307.
    1. Accurso V, Santoro M, Raso S, et al. . Splenomegaly impacts prognosis in essential thrombocythemia and polycythemia vera: a single center study. Hematol Rep. 2019;11(4):8281.
    1. Radia D, Geyer HL. Management of symptoms in polycythemia vera and essential thrombocythemia patients. Hematology (Am Soc Hematol Educ Program). 2015;2015(1):340-348.
    1. Mesa RA, Schwager S, Radia D, et al. . The Myelofibrosis Symptom Assessment Form (MFSAF): an evidence-based brief inventory to measure quality of life and symptomatic response to treatment in myelofibrosis. Leuk Res. 2009;33(9):1199-1203.
    1. Tefferi A, Rumi E, Finazzi G, et al. . Survival and prognosis among 1545 patients with contemporary polycythemia vera: an international study. Leukemia. 2013;27(9):1874-1881.
    1. Konopleva M, Martinelli G, Daver N, et al. . MDM2 inhibition: an important step forward in cancer therapy. Leukemia. 2020;34(11):2858-2874.
    1. Momand J, Jung D, Wilczynski S, Niland J. The MDM2 gene amplification database. Nucleic Acids Res. 1998;26(15):3453-3459.
    1. Yee K, Papayannidis C, Vey N, et al. . Murine double minute 2 inhibition alone or with cytarabine in acute myeloid leukemia: results from an idasanutlin phase 1/1b study. Leuk Res. 2021;100:106489.
    1. Lu M, Wang X, Li Y, et al. . Combination treatment in vitro with nutlin, a small-molecule antagonist of MDM2, and pegylated interferon-α 2a specifically targets JAK2V617F-positive polycythemia vera cells. Blood. 2012;120(15):3098-3105.
    1. Mascarenhas J, Lu M, Kosiorek H, et al. . Oral idasanutlin in patients with polycythemia vera. Blood. 2019;134(6):525-533.
    1. Arber DA, Orazi A, Hasserjian R, et al. . The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia [published correction appears in Blood. 2016;128(3):46–463]. Blood. 2016;127(20):2391-2405.
    1. Barosi G, Birgegard G, Finazzi G, et al. . A unified definition of clinical resistance and intolerance to hydroxycarbamide in polycythaemia vera and primary myelofibrosis: results of a European LeukemiaNet (ELN) consensus process. Br J Haematol. 2010;148(6):961-963.
    1. Barbui T, Barosi G, Birgegard G, et al. ; European LeukemiaNet . Philadelphia-negative classical myeloproliferative neoplasms: critical concepts and management recommendations from European LeukemiaNet. J Clin Oncol. 2011;29(6):761-770.
    1. Barosi G, Birgegard G, Finazzi G, et al. . Response criteria for essential thrombocythemia and polycythemia vera: result of a European LeukemiaNet consensus conference. Blood. 2009;113(20):4829-4833.
    1. Barosi G, Mesa R, Finazzi G, et al. . Revised response criteria for polycythemia vera and essential thrombocythemia: an ELN and IWG-MRT consensus project. Blood. 2013;121(23):4778-4781.
    1. Abelsson J, Andréasson B, Samuelsson J, et al. . Patients with polycythemia vera have worst impairment of quality of life among patients with newly diagnosed myeloproliferative neoplasms. Leuk Lymphoma. 2013;54(10):2226-2230.
    1. Emanuel RM, Dueck AC, Geyer HL, et al. . Myeloproliferative neoplasm (MPN) symptom assessment form total symptom score: prospective international assessment of an abbreviated symptom burden scoring system among patients with MPNs [published correction appears in J Clin Oncol. 2012;30(36):4590]. J Clin Oncol. 2012;30(33):4098-4103.
    1. Scherber R, Dueck AC, Johansson P, et al. . The Myeloproliferative Neoplasm Symptom Assessment Form (MPN-SAF): international prospective validation and reliability trial in 402 patients. Blood. 2011;118(2):401-408.
    1. Aaronson NK, Ahmedzai S, Bergman B, et al. . The European Organization for Research and Treatment of Cancer QLQ-C30: a quality-of-life instrument for use in international clinical trials in oncology. J Natl Cancer Inst. 1993;85(5):365-376.
    1. Fitzsimmons D, Johnson CD, George S, et al. ; EORTC Study Group on Quality of Life . Development of a disease specific quality of life (QoL) questionnaire module to supplement the EORTC core cancer QoL questionnaire, the QLQ-C30 in patients with pancreatic cancer. Eur J Cancer. 1999;35(6):939-941.
    1. Mesa R, Verstovsek S, Kiladjian JJ, et al. . Changes in quality of life and disease-related symptoms in patients with polycythemia vera receiving ruxolitinib or standard therapy. Eur J Haematol. 2016;97(2):192-200.
    1. FoundationOneHeme. Technical specifications. . Accessed 15 June 2021.
    1. Sun JX, He Y, Sanford E, et al. . A computational approach to distinguish somatic vs. germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal. PLOS Comput Biol. 2018;14(2):e1005965.
    1. National Cancer Center Network. Myeloproliferative neoplasms (version 1.2020). . Accessed 8 March 2021.
    1. Alvarez-Larrán A, Pereira A, Cervantes F, et al. . Assessment and prognostic value of the European LeukemiaNet criteria for clinicohematologic response, resistance, and intolerance to hydroxyurea in polycythemia vera. Blood. 2012;119(6):1363-1369.
    1. Assi TB, Baz E. Current applications of therapeutic phlebotomy. Blood Transfus. 2014;12(suppl 1):s75-s83.
    1. Kiladjian JJ, Chomienne C, Fenaux P. Interferon-alpha therapy in bcr-abl-negative myeloproliferative neoplasms. Leukemia. 2008;22(11):1990-1998.
    1. Sever M, Newberry KJ, Verstovsek S. Therapeutic options for patients with polycythemia vera and essential thrombocythemia refractory/resistant to hydroxyurea. Leuk Lymphoma. 2014;55(12):2685-2690.
    1. Vannucchi AM, Kiladjian JJ, Griesshammer M, et al. . Ruxolitinib versus standard therapy for the treatment of polycythemia vera. N Engl J Med. 2015;372(5):426-435.
    1. Nazha A, Khoury JD, Verstovsek S, Daver N. Second line therapies in polycythemia vera: what is the optimal strategy after hydroxyurea failure? Crit Rev Oncol Hematol. 2016;105:112-117.
    1. Marchioli R, Finazzi G, Specchia G, et al. ; CYTO-PV Collaborative Group . Cardiovascular events and intensity of treatment in polycythemia vera. N Engl J Med. 2013;368(1):22-33.
    1. Passamonti F, Griesshammer M, Palandri F, et al. . Ruxolitinib for the treatment of inadequately controlled polycythaemia vera without splenomegaly (RESPONSE-2): a randomised, open-label, phase 3b study. Lancet Oncol. 2017;18(1):88-99.
    1. Ronner L, Podoltsev N, Gotlib J, et al. . Persistent leukocytosis in polycythemia vera is associated with disease evolution but not thrombosis. Blood. 2020;135(19):1696-1703.
    1. Colafigli G, Scalzulli E, Pepe S, et al. . The advantages and risks of ruxolitinib for the treatment of polycythemia vera. Expert Rev Hematol. 2020; 13(10):1067-1072.
    1. Kiladjian JJ, Zachee P, Hino M, et al. . Long-term efficacy and safety of ruxolitinib versus best available therapy in polycythaemia vera (RESPONSE): 5-year follow up of a phase 3 study. Lancet Haematol. 2020;7(3):e226-e237.
    1. Yacoub A, Mascarenhas J, Kosiorek H, et al. . Pegylated interferon alfa-2a for polycythemia vera or essential thrombocythemia resistant or intolerant to hydroxyurea. Blood. 2019;134(18):1498-1509.
    1. Gisslinger H, Klade C, Georgiev P, et al. ; PROUD-PV Study Group . Ropeginterferon alfa-2b versus standard therapy for polycythaemia vera (PROUD-PV and CONTINUATION-PV): a randomised, non-inferiority, phase 3 trial and its extension study. Lancet Haematol. 2020;7(3):e196-e208.
    1. Masarova L, Patel KP, Newberry KJ, et al. . Pegylated interferon alfa-2a in patients with essential thrombocythaemia or polycythaemia vera: a post-hoc, median 83 month follow-up of an open-label, phase 2 trial. Lancet Haematol. 2017;4(4):e165-e175.
    1. Vannucchi AM, Pieri L, Guglielmelli P. JAK2 allele burden in the myeloproliferative neoplasms: effects on phenotype, prognosis and change with treatment. Ther Adv Hematol. 2011;2(1):21-32.
    1. Antonioli E, Carobbio A, Pieri L, et al. . Hydroxyurea does not appreciably reduce JAK2 V617F allele burden in patients with polycythemia vera or essential thrombocythemia. Haematologica. 2010;95(8):1435-1438.
    1. Vannucchi AM, Antonioli E, Guglielmelli P, et al. ; MPD Research Consortium . Prospective identification of high-risk polycythemia vera patients based on JAK2(V617F) allele burden. Leukemia. 2007;21(9):1952-1959.
    1. Alimam S, Harrison C. Experience with ruxolitinib in the treatment of polycythaemia vera [published correction appears in Ther Adv Hematol. 2017;8(9):273]. Ther Adv Hematol. 2017;8(4):139-151.
    1. Shadfan M, Lopez-Pajares V, Yuan ZM. MDM2 and MDMX: alone and together in regulation of p53. Transl Cancer Res. 2012;1(2):88-89.
    1. Marcellino BK, Farnoud N, Cassinat B, et al. . Transient expansion of TP53 mutated clones in polycythemia vera patients treated with idasanutlin. Blood Adv. 2020;4(22):5735-5744.
    1. Grinfeld J, Nangalia J, Baxter EJ, et al. . Classification and personalized prognosis in myeloproliferative neoplasms. N Engl J Med. 2018;379(15):1416-1430.
    1. Sallman DA, McLemore AF, Aldrich AL, et al. . TP53 mutations in myelodysplastic syndromes and secondary AML confer an immunosuppressive phenotype. Blood. 2020;136(24):2812-2823.
    1. Gotlib J, Gabrail N, O’Connell CL, et al. . A randomized, open-label, multicenter, phase 2 study to evaluate the efficacy, safety, and pharmacokinetics of KRT-232 compared with ruxolitinib in patients with phlebotomy-dependent polycythemia vera. Blood. 2019;134(suppl 1):4168.

Source: PubMed

3
購読する