A Phase I/II Open-Label Study of Molibresib for the Treatment of Relapsed/Refractory Hematologic Malignancies

Mark A Dawson, Gautam Borthakur, Brian J P Huntly, Anastasios Karadimitris, Adrian Alegre, Aristeidis Chaidos, Dan T Vogl, Daniel A Pollyea, Faith E Davies, Gareth J Morgan, Jacob L Glass, Manali Kamdar, Maria-Victoria Mateos, Natalia Tovar, Paul Yeh, Regina García Delgado, Faisal Basheer, Ludovica Marando, Paolo Gallipoli, Anastasia Wyce, Anu Shilpa Krishnatry, Olena Barbash, Evi Bakirtzi, Geraldine Ferron-Brady, Natalie O Karpinich, Michael T McCabe, Shawn W Foley, Thierry Horner, Arindam Dhar, Brandon E Kremer, Michael Dickinson, Mark A Dawson, Gautam Borthakur, Brian J P Huntly, Anastasios Karadimitris, Adrian Alegre, Aristeidis Chaidos, Dan T Vogl, Daniel A Pollyea, Faith E Davies, Gareth J Morgan, Jacob L Glass, Manali Kamdar, Maria-Victoria Mateos, Natalia Tovar, Paul Yeh, Regina García Delgado, Faisal Basheer, Ludovica Marando, Paolo Gallipoli, Anastasia Wyce, Anu Shilpa Krishnatry, Olena Barbash, Evi Bakirtzi, Geraldine Ferron-Brady, Natalie O Karpinich, Michael T McCabe, Shawn W Foley, Thierry Horner, Arindam Dhar, Brandon E Kremer, Michael Dickinson

Abstract

Purpose: Molibresib is a selective, small molecule inhibitor of the bromodomain and extra-terminal (BET) protein family. This was an open-label, two-part, Phase I/II study investigating molibresib monotherapy for the treatment of hematological malignancies (NCT01943851).

Patients and methods: Part 1 (dose escalation) determined the recommended Phase 2 dose (RP2D) of molibresib in patients with acute myeloid leukemia (AML), Non-Hodgkin lymphoma (NHL), or multiple myeloma. Part 2 (dose expansion) investigated the safety and efficacy of molibresib at the RP2D in patients with relapsed/refractory myelodysplastic syndrome (MDS; as well as AML evolved from antecedent MDS) or cutaneous T-cell lymphoma (CTCL). The primary endpoint in Part 1 was safety and the primary endpoint in Part 2 was objective response rate (ORR).

Results: There were 111 patients enrolled (87 in Part 1, 24 in Part 2). Molibresib RP2Ds of 75 mg daily (for MDS) and 60 mg daily (for CTCL) were selected. Most common Grade 3+ adverse events included thrombocytopenia (37%), anemia (15%), and febrile neutropenia (15%). Six patients achieved complete responses [3 in Part 1 (2 AML, 1 NHL), 3 in Part 2 (MDS)], and 7 patients achieved partial responses [6 in Part 1 (4 AML, 2 NHL), 1 in Part 2 (MDS)]. The ORRs for Part 1, Part 2, and the total study population were 10% [95% confidence interval (CI), 4.8-18.7], 25% (95% CI, 7.3-52.4), and 13% (95% CI, 6.9-20.6), respectively.

Conclusions: While antitumor activity was observed with molibresib, use was limited by gastrointestinal and thrombocytopenia toxicities. Investigations of molibresib as part of combination regimens may be warranted.

©2022 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
A, Serial bone marrow blasts and mutation burden during molibresib therapy (60 mg QD). There was an initial rise in bone marrow blasts and mutational burden in IDH1 Arg132Cys and TP53 Asp259Tyr during molibresib treatment, but these decreased by day 98. B, Serial bone marrow trephine assessments showing a reduction in blasts by H&E stain (top) and by CD34 IHC (bottom), as well as evidence of regenerative erythropoiesis at day 98 of molibresib treatment. H&E, hematoxylin and eosin.
Figure 2.
Figure 2.
A, RT-qPCR to measure MYC expression was performed on bone marrow aspirate samples collected at screening and post-molibresib treatment. Bars represent the log2-fold change of MYC expression after treatment relative to screening. B, Volcano plot graphing the log10(FDR) versus the log2(fold change) for 7,978 genes that are expressed (>3 FPKM) in all analyzed samples. Blue dots indicate genes that pass the significance threshold of an FDR <0.05 and >1.5-fold change, whereas pink dots do not meet both criteria. C, A heatmap of all genes that are significantly differentially expressed when pooled across all 13 patients. Each column corresponds to the log2(fold change) for each patient. The bar at the top of the heatmap corresponds to clinical response, indicating CR (pink), PR (blue), or NR (green). D, GSEA results when examining the average fold change across all 13 patients. Twelve gene sets were significantly downregulated (FDR <0.05). Two example enrichment plots show the enrichment score (black line) across all genes, with black bars, indicating genes within the interferon alpha response and MYC target v2 gene sets, respectively. AML, acute myeloid leukemia; CR, complete response; FC, fold change; FDR, false discovery rate; FPKM, fragments per kilobase per million reads; GSEA, gene set enrichment analysis; NR, non-responder; PR, partial response; RT-qPCR, reverse transcription and quantitative polymerase chain reaction.
Figure 3.
Figure 3.
A and B, Heatmaps of individual patients’ change in gene expression following molibresib treatment (log2; fold change) for genes that were identified as differentially expressed when pooled across all NRs (A), or a clinical response (CR and PR; B). Genes are defined as differentially expressed if they reach a 1.5-fold change with an FDR <0.05. The bar at the top of the heatmap corresponds to clinical response, indicating CR [pink; Patient 1 with CRp, Patient 2 with CRi), PR (blue), or NR (green)]. C, Venn diagram showing the number of differentially expressed genes in responders, non-responders, or both. D, Scatterplot showing the fold change in gene expression for the 2 patients who achieved CR. Genes are categorized as unchanged in both patients (gray), differentially expressed in one patient (blue), differentially expressed in the same direction in both patients (black), or differentially expressed in both patients, but in the opposite direction (red) if they exhibited >1.5-fold change (dotted lines). CR, complete response; CRi, CR but platelet count <100×109/L or neutrophil count <1×109/L; CRp, CR but platelet count <100×109/L; FDR, false discovery rate; NR, non-responder; PR, partial response.

References

    1. French CA. Small-molecule targeting of BET proteins in cancer. Adv Cancer Res 2016;131:21–58.
    1. Mujtaba S, Zeng L, Zhou MM. Structure and acetyl-lysine recognition of the bromodomain. Oncogene 2007;26:5521–7.
    1. Muller S, Filippakopoulos P, Knapp S. Bromodomains as therapeutic targets. Expert Rev Mol Med 2011;13:e29.
    1. Filippakopoulos P, Picaud S, Mangos M, Keates T, Lambert JP, Barsyte-Lovejoy D, et al. . Histone recognition and large-scale structural analysis of the human bromodomain family. Cell 2012;149:214–31.
    1. Dey A, Nishiyama A, Karpova T, McNally J, Ozato K. Brd4 marks select genes on mitotic chromatin and directs postmitotic transcription. Mol Biol Cell 2009;20:4899–909.
    1. Yang Z, He N, Zhou Q. Brd4 recruits P-TEFb to chromosomes at late mitosis to promote G1 gene expression and cell cycle progression. Mol Cell Biol 2008;28:967–76.
    1. Dawson MA. The cancer epigenome: concepts, challenges, and therapeutic opportunities. Science 2017;355:1147–52.
    1. Manzotti G, Ciarrocchi A, Sancisi V. Inhibition of BET proteins and histone deacetylase (HDACs): crossing roads in cancer therapy. Cancers 2019;11:304.
    1. Belkina AC, Denis GV. BET domain co-regulators in obesity, inflammation and cancer. Nat Rev Cancer 2012;12:465–77.
    1. Padmanabhan B, Mathur S, Manjula R, Tripathi S. Bromodomain and extra-terminal (BET) family proteins: new therapeutic targets in major diseases. J Biosci 2016;41:295–311.
    1. Dawson MA, Kouzarides T, Huntly BJ. Targeting epigenetic readers in cancer. N Engl J Med 2012;367:647–57.
    1. Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, et al. . BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 2011;146:904–17.
    1. Dawson MA, Prinjha RK, Dittmann A, Giotopoulos G, Bantscheff M, Chan WI, et al. . Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 2011;478:529–33.
    1. Zuber J, Shi J, Wang E, Rappaport AR, Herrmann H, Sison EA, et al. . RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature 2011;478:524–8.
    1. Gabay M, Li Y, Felsher DW. MYC activation is a hallmark of cancer initiation and maintenance. Cold Spring Harb Perspect Med 2014;4:a014241.
    1. Perez-Salvia M, Esteller M. Bromodomain inhibitors and cancer therapy: from structures to applications. Epigenetics 2017;12:323–39.
    1. Nicodeme E, Jeffrey KL, Schaefer U, Beinke S, Dewell S, Chung CW, et al. . Suppression of inflammation by a synthetic histone mimic. Nature 2010;468:1119–23.
    1. Piha-Paul SA, Hann CL, French CA, Cousin S, Braña I, Cassier PA, et al. . Phase 1 study of molibresib (GSK525762), a bromodomain and extra-terminal domain protein inhibitor, in NUT carcinoma and other solid tumors. JNCI Canc Spectr 2019;4:pkz093.
    1. Cousin S, Blay JY, Brana Garcia I, de Bono JS, Le Tourneau C, Moreno V, et al. . Safety, pharmacokinetic, pharmacodynamic and clinical activity of molibresib for the treatment of NUT carcinoma and other cancers: results of a phase I/II open-label, dose escalation study. Int J Cancer 2022;150:993–1006.
    1. Riddell K, Patel A, Collins G, Zhou Y, Schramek D, Kremer BE, et al. . An adaptive physiologically based pharmacokinetic-driven design to investigate the effect of itraconazole and rifampicin on the pharmacokinetics of molibresib (GSK525762) in healthy female volunteers. J Clin Pharmacol 2021;61:125–37.
    1. Krishnatry AS, Voelkner A, Dhar A, Prohn M, Ferron-Brady G. Population pharmacokinetic modeling of molibresib and its active metabolites in patients with solid tumors: a semimechanistic autoinduction model. CPT Pharmacometrics Syst Pharmacol 2021;10:709–22.
    1. Bell CC, Fennell KA, Chan YC, Rambow F, Yeung MM, Vassiliadis D, et al. . Targeting enhancer switching overcomes non-genetic drug resistance in acute myeloid leukaemia. Nat Commun 2019;10:2723.
    1. Wyce A, Matteo JJ, Foley SW, Felitsky DJ, Rajapurkar SR, Zhang XP, et al. . MEK inhibitors overcome resistance to BET inhibition across a number of solid and hematologic cancers. Oncogenesis 2018;7:35.
    1. Dawson MA, Gudgin EJ, Horton SJ, Giotopoulos G, Meduri E, Robson S, et al. . Recurrent mutations, including NPM1c, activate a BRD4-dependent core transcriptional program in acute myeloid leukemia. Leukemia 2014;28:311–20.
    1. Chaidos A, Caputo V, Gouvedenou K, Liu B, Marigo I, Chaudhry MS, et al. . Potent antimyeloma activity of the novel bromodomain inhibitors I-BET151 and I-BET762. Blood 2014;123:697–705.
    1. Neuenschwander B, Branson M, Gsponer T. Critical aspects of the Bayesian approach to phase I cancer trials. Stat Med 2008;27:2420–39.
    1. Cheson BD, Greenberg PL, Bennett JM, Lowenberg B, Wijermans PW, Nimer SD, et al. . Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia. Blood 2006;108:419–25.
    1. Cheson BD, Bennett JM, Kopecky KJ, Buchner T, Willman CL, Estey EH, et al. . Revised recommendations of the International Working Group for diagnosis, standardization of response criteria, treatment outcomes, and reporting standards for therapeutic trials in acute myeloid leukemia. J Clin Oncol 2003;21:4642–9.
    1. Olsen EA, Whittaker S, Kim YH, Duvic M, Prince HM, Lessin SR, et al. . Clinical end points and response criteria in mycosis fungoides and Sezary syndrome: a consensus statement of the International Society for Cutaneous Lymphomas, the United States Cutaneous Lymphoma Consortium, and the Cutaneous Lymphoma Task Force of the European Organisation for Research and Treatment of Cancer. J Clin Oncol 2011;29:2598–607.
    1. Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, Tamayo P. The molecular signatures database (MSigDB) hallmark gene set collection. Cell Syst 2015;1:417–25.
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. . Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 2005;102:15545–50.
    1. Broad Institute Inc. GSEA: gene set enrichment analysis. [Accessed 2021 Dec 1]. Available from:.
    1. Gregory RW, Ben B, Lodewijk B, Robert G, Wolfgang H, Andy L, et al. . Various R programming tools for plotting data. R package version 3.0.1. Current version (3.1.1). 2016. [Accessed 2021 Sep 12]. Available from:.
    1. Green SJ, Dahlberg S. Planned versus attained design in phase II clinical trials. Stat Med 1992;11:853–62.
    1. Fennell KA, Bell CC, Dawson MA. Epigenetic therapies in acute myeloid leukemia: where to from here? Blood 2019;134:1891–901.
    1. Morel D, Jeffery D, Aspeslagh S, Almouzni G, Postel-Vinay S. Combining epigenetic drugs with other therapies for solid tumours—past lessons and future promise. Nat Rev Clin Oncol 2020;17:91–107.
    1. Stein EM, Garcia-Manero G, Rizzieri DA, Tibes R, Berdeja JG, Savona MR, et al. . The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia. Blood 2018;131:2661–9.
    1. Salamero O, Montesinos P, Willekens C, Perez-Simon JA, Pigneux A, Recher C, et al. . First-in-human phase I study of iadademstat (ORY-1001): a first-in-class lysine-specific histone demethylase 1A inhibitor, in relapsed or refractory acute myeloid leukemia. J Clin Oncol 2020;38:4260–73.
    1. . Vorinostat dose-escalation after allogeneic hematopoietic cell transplantation. 2019. Available from:
    1. . Epigenetic reprogramming in relapse/refractory AML. 2017. Available from:
    1. . Nivolumab in combination with 5-azacytidine in childhood relapsed/refractory AML. 2019. Available from: .
    1. . Pracinostat and gemtuzumab ozogamicin (PraGO) in patients with relapsed/refractory acute myeloid leukemia. 2019. Available from:
    1. Wass M, Gollner S, Besenbeck B, Schlenk RF, Mundmann P, Gothert JR, et al. . A proof-of-concept phase I/II pilot trial of LSD1 inhibition by tranylcypromine combined with ATRA in refractory/relapsed AML patients not eligible for intensive therapy. Leukemia 2021;35:701–11.

Source: PubMed

3
Abonneren