Phase 1 Study of Molibresib (GSK525762), a Bromodomain and Extra-Terminal Domain Protein Inhibitor, in NUT Carcinoma and Other Solid Tumors

Sarina A Piha-Paul, Christine L Hann, Christopher A French, Sophie Cousin, Irene Braña, Phillippe A Cassier, Victor Moreno, Johann S de Bono, Sara Duckworth Harward, Geraldine Ferron-Brady, Olena Barbash, Anastasia Wyce, Yuehui Wu, Thierry Horner, Meg Annan, Nigel J Parr, Rabinder K Prinjha, Christopher L Carpenter, John Hilton, David S Hong, Naomi B Haas, Mark C Markowski, Arindam Dhar, Peter J O'Dwyer, Geoffrey I Shapiro, Sarina A Piha-Paul, Christine L Hann, Christopher A French, Sophie Cousin, Irene Braña, Phillippe A Cassier, Victor Moreno, Johann S de Bono, Sara Duckworth Harward, Geraldine Ferron-Brady, Olena Barbash, Anastasia Wyce, Yuehui Wu, Thierry Horner, Meg Annan, Nigel J Parr, Rabinder K Prinjha, Christopher L Carpenter, John Hilton, David S Hong, Naomi B Haas, Mark C Markowski, Arindam Dhar, Peter J O'Dwyer, Geoffrey I Shapiro

Abstract

Background: Bromodomain and extra-terminal domain proteins are promising epigenetic anticancer drug targets. This first-in-human study evaluated the safety, recommended phase II dose, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity of the bromodomain and extra-terminal domain inhibitor molibresib (GSK525762) in patients with nuclear protein in testis (NUT) carcinoma (NC) and other solid tumors.

Methods: This was a phase I and II, open-label, dose-escalation study. Molibresib was administered orally once daily. Single-patient dose escalation (from 2 mg/d) was conducted until the first instance of grade 2 or higher drug-related toxicity, followed by a 3 + 3 design. Pharmacokinetic parameters were obtained during weeks 1 and 3. Circulating monocyte chemoattractant protein-1 levels were measured as a pharmacodynamic biomarker.

Results: Sixty-five patients received molibresib. During dose escalation, 11% experienced dose-limiting toxicities, including six instances of grade 4 thrombocytopenia, all with molibresib 60-100 mg. The most frequent treatment-related adverse events of any grade were thrombocytopenia (51%) and gastrointestinal events, including nausea, vomiting, diarrhea, decreased appetite, and dysgeusia (22%-42%), anemia (22%), and fatigue (20%). Molibresib demonstrated an acceptable safety profile up to 100 mg; 80 mg once daily was selected as the recommended phase II dose. Following single and repeat dosing, molibresib showed rapid absorption and elimination (maximum plasma concentration: 2 hours; t1/2: 3-7 hours). Dose-dependent reductions in circulating monocyte chemoattractant protein-1 levels were observed. Among 19 patients with NC, four achieved either confirmed or unconfirmed partial response, eight had stable disease as best response, and four were progression-free for more than 6 months.

Conclusions: Once-daily molibresib was tolerated at doses demonstrating target engagement. Preliminary data indicate proof-of-concept in NC.

© The Author(s) 2019. Published by Oxford University Press.

Figures

Figure 1.
Figure 1.
Pharmacokinetic parameters and pharmacodynamic profiles of molibresib. A) Pharmacokinetic profiles of molibresib (80-mg dose) and its major active human metabolites M5 and M13 measured together at week 1 day 1 (left) and week 3 day 4 (right). B) Maximum change in platelet count from baseline (%) by molibresib dose (left) by exposure measured on week 1 day 1 (right). Platelet data are for the initial dose level during the first 60 days on treatment and limited to patients who received the planned first 2 weeks of treatment. C) Percent maximum change in Factor VII from baseline by molibresib dose at week 1 (left) and week 3 (right). D) Percent maximum change in MCP-1 from baseline by molibresib dose at week 1. AUC(0–24) = area under the concentration–time curve from 0 to 24 hours; MCP-1 = monocyte chemoattractant protein 1. In B–D, predicted values represent the line of best fit (ie, a simple maximum effect [Emax] model) through observed values.
Figure 2.
Figure 2.
Antitumor activity of molibresib. A) Maximum tumor reduction (% from baseline) and best clinical responses for all treated patients who had measurable disease and at least one complete postbaseline assessment (n = 46 of 65 enrolled). B) Maximum tumor reduction (% from baseline) and best clinical responses for NC patients who had measurable disease and at least one complete postbaseline assessment (n = 14 of 19 enrolled). In A and B, NE indicates patients with clinical progression, with the postbaseline scan performed before 28 days from baseline. Patients not depicted in A and B were removed from the trial for early clinical progression or progression based on evidence of new lesion(s) without reassessment of baseline target lesions or had nonmeasurable disease at outset. C) Progression-free survival (PFS) in the NC cohort for patients who received at least 60 mg daily (n = 11). Tick marks represent censored patients: If a patient received subsequent anticancer therapy before the date of documented events, PFS was censored at the last adequate assessment before the initiation of therapy. Otherwise, if a patient did not have a documented date of events, PFS was censored at the date of the last adequate assessment. aMaximum reduction from baseline is 0%. bRemained on treatment for greater than 4 months. cConfirmed response. CRC = colorectal cancer; NC = NUT carcinoma; NSCLC = non-small cell lung cancer; NUT = nuclear protein in testis; SCLC = small cell lung cancer; PD = progressive disease; PR = partial response; QD = once daily; SD = stable disease.

References

    1. Mujtaba S, Zeng L, Zhou MM.. Structure and acetyl-lysine recognition of the bromodomain. Oncogene. 2007;26(37):5521–5527.
    1. French CA. Small-molecule targeting of BET proteins in cancer. Adv Cancer Res. 2016;131:21–58.
    1. Dey A, Nishiyama A, Karpova T, et al. Brd4 marks select genes on mitotic chromatin and directs postmitotic transcription. Mol Biol Cell. 2009;20(23):4899–4909.
    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(3):967–976.
    1. Belkina AC, Denis GV.. BET domain co-regulators in obesity, inflammation and cancer. Nat Rev Cancer. 2012;12(7):465–477.
    1. Padmanabhan B, Mathur S, Manjula R, et al. Bromodomain and extra-terminal (BET) family proteins: new therapeutic targets in major diseases. J Biosci. 2016;41(2):295–311.
    1. Perez-Salvia M, Esteller M.. Bromodomain inhibitors and cancer therapy: from structures to applications. Epigenetics. 2017;12(5):323–339.
    1. Grayson AR, Walsh EM, Cameron MJ, et al. MYC, a downstream target of BRD-NUT, is necessary and sufficient for the blockade of differentiation in NUT midline carcinoma. Oncogene. 2014;33(13):1736–1742.
    1. Bauer DE, Mitchell CM, Strait KM, et al. Clinicopathologic features and long-term outcomes of NUT midline carcinoma. Clin Cancer Res. 2012;18(20):5773–5779.
    1. Chau NG, Hurwitz S, Mitchell CM, et al. Intensive treatment and survival outcomes in NUT midline carcinoma of the head and neck. Cancer. 2016;122(23):3632–3640.
    1. French CA, Ramirez CL, Kolmakova J, et al. BRD-NUT oncoproteins: a family of closely related nuclear proteins that block epithelial differentiation and maintain the growth of carcinoma cells. Oncogene. 2008;27(15):2237–2242.
    1. Alekseyenko AA, Walsh EM, Wang X, et al. The oncogenic BRD4-NUT chromatin regulator drives aberrant transcription within large topological domains. Genes Dev. 2015;29(14):1507–1523.
    1. Delmore JE, Issa GC, Lemieux ME, et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell. 2011;146(6):904–917.
    1. Mertz JA, Conery AR, Bryant BM, et al. Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc Natl Acad Sci USA. 2011;108(40):16669–16674.
    1. Perez-Salvia M, Simo-Riudalbas L, Llinas-Arias P, et al. Bromodomain inhibition shows antitumoral activity in mice and human luminal breast cancer. Oncotarget. 2017;8(31):51621–51629.
    1. Zhao Y, Yang CY, Wang S.. The making of I-BET762, a BET bromodomain inhibitor now in clinical development. J Med Chem. 2013;56(19):7498–7500.
    1. Chaidos A, Caputo V, Gouvedenou K, et al. Potent antimyeloma activity of the novel bromodomain inhibitors I-BET151 and I-BET762. Blood. 2014;123(5):697–705.
    1. Wyce A, Degenhardt Y, Bai Y, et al. Inhibition of BET bromodomain proteins as a therapeutic approach in prostate cancer. Oncotarget. 2013;4(12):2419–2429.
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–247.
    1. National Cancer Institute. Common Terminology Criteria for Adverse Events v4.0, 2009. . Accessed August 18, 2019.
    1. Belkina AC, Nikolajczyk BS, Denis GV.. BET protein function is required for inflammation: Brd2 genetic disruption and BET inhibitor JQ1 impair mouse macrophage inflammatory responses. J Immunol. 2013;190(7):3670–3678.
    1. Nguyen TH, Maltby S, Eyers F, et al. Bromodomain and extra terminal (BET) inhibitor suppresses macrophage-driven steroid-resistant exacerbations of airway hyper-responsiveness and inflammation. PLoS One. 2016;11(9):e0163392..
    1. Scher HI, Halabi S, Tannock I, et al. Design and end points of clinical trials for patients with progressive prostate cancer and castrate levels of testosterone: recommendations of the Prostate Cancer Clinical Trials Working Group. J Clin Oncol. 2008;26(7):1148–1159.
    1. Doroshow DB, Eder JP, LoRusso PM.. BET inhibitors: a novel epigenetic approach. Ann Oncol. 2017;28(8):1776–1787.
    1. Lewin J, Soria JC, Stathis A, et al. Phase Ib trial with birabresib, a small-molecule inhibitor of bromodomain and extraterminal proteins, in patients with selected advanced solid tumors. J Clin Oncol. 2018;36(30):3007–3014.
    1. Chau NG, Clement M, Danga K, et al. An anatomical site and genetic based prognostic model for patients with NUT midline carcinoma: analysis of 124 patients. JNCI Cancer Spectr. doi: 10.1093/jncics/pkz094.
    1. Hilton J, Cristea MC, Voskoboynik M, et al. 4110: initial results from a phase I/IIa trial evaluating BMS-986158, an inhibitor of the bromodomain and extra-terminal (BET) proteins, in patients (pts) with advanced cancer. Ann Oncol. 2018;29(suppl 8):viii134.
    1. Shapiro GI, Dowlati A, LoRusso PM, et al. Clinically efficacy of the BET bromodomain inhibitor TEN-010 in an open-label substudy with patients with documented NUT-midline carcinoma (NMC). Mol Cancer Ther. 2015;14(12 suppl 2):A49.
    1. Yang L, Zhang Y, Shan W, et al. Repression of BET activity sensitizes homologous recombination-proficient cancers to PARP inhibition. Sci Transl Med. 2017;9(400):eaal1645.
    1. Zhang J, Dulak AM, Hattersley MM, et al. BRD4 facilitates replication stress-induced DNA damage response. Oncogene. 2018;37(28):3763–3777.
    1. Stathis A, Zucca E, Bekradda M, et al. Clinical response of carcinomas harboring the BRD4-NUT oncoprotein to the targeted bromodomain inhibitor OTX015/MK-8628. Cancer Discov. 2016;6(5):492–500.
    1. Shu S, Polyak K.. BET bromodomain proteins as cancer therapeutic targets. Cold Spring Harb Symp Quant Biol. 2016;81:123–129.
    1. Amorim S, Stathis A, Gleeson M, et al. Bromodomain inhibitor OTX015 in patients with lymphoma or multiple myeloma: a dose-escalation, open-label, pharmacokinetic, phase 1 study. Lancet Haematol. 2016;3(4):e196–e204.
    1. Dawson M, Stein EM, Huntly BJP, et al. A phase I study of GSK525762, a selective bromodomain (BRD) and extra terminal protein (BET) inhibitor: results from part 1 of phase I/II open label single agent study in patients with acute myeloid leukemia (AML). Blood. 2017;130(Suppl 1):Abstract 1377.
    1. Sparano JA, Cescon DW, Oliveira M, et al. A phase I/II dose escalation and expansion study to investigate the safety, pharmacokinetics, pharmacodynamics and clinical activity of GSK525762 in combination with fulvestrant in subjects with ER+ breast cancer. J Clin Oncol. 2017;35(Suppl 1):Abstract TPS 1114.
    1. Vaishampayan UN, Narayan V, Wise D, et al. A phase Ib open-label, dose escalation and expansion study to investigate the safety, pharmacokinetics, pharmacodynamics and clinical activity of GSK525762 in combination with abiraterone or enzalutamide in metastatic castrate-resistant prostate cancer. J Clin Oncol. 2018;36(suppl):Abstract TPS391.

Source: PubMed

3
S'abonner