A Phase 1 study of RO6870810, a novel bromodomain and extra-terminal protein inhibitor, in patients with NUT carcinoma, other solid tumours, or diffuse large B-cell lymphoma

Geoffrey I Shapiro, Patricia LoRusso, Afshin Dowlati, Khanh T Do, Caron A Jacobson, Ulka Vaishampayan, Amy Weise, Paolo F Caimi, Joseph Paul Eder, Christopher A French, Emily Labriola-Tompkins, Frédéric Boisserie, William E Pierceall, Jianguo Zhi, Sharon Passe, Mark DeMario, Martin Kornacker, Philippe Armand, Geoffrey I Shapiro, Patricia LoRusso, Afshin Dowlati, Khanh T Do, Caron A Jacobson, Ulka Vaishampayan, Amy Weise, Paolo F Caimi, Joseph Paul Eder, Christopher A French, Emily Labriola-Tompkins, Frédéric Boisserie, William E Pierceall, Jianguo Zhi, Sharon Passe, Mark DeMario, Martin Kornacker, Philippe Armand

Abstract

Background: Bromodomain and extra-terminal (BET) proteins are epigenetic readers that can drive carcinogenesis and therapy resistance. RO6870810 is a novel, small-molecule BET inhibitor.

Methods: We conducted a Phase 1 study of RO6870810 administered subcutaneously for 21 or 14 days of 28- or 21-day cycles, respectively, in patients with the nuclear protein of the testis carcinoma (NC), other solid tumours, or diffuse large B-cell lymphoma (DLBCL) with MYC deregulation.

Results: Fatigue (42%), decreased appetite (35%) and injection-site erythema (35%) were the most common treatment-related adverse events. Pharmacokinetic parameters demonstrated linearity over the dose range tested and support once-daily dosing. Pharmacodynamic assessments demonstrated sustained decreases in CD11b levels in peripheral blood mononuclear cells. Objective response rates were 25% (2/8), 2% (1/47) and 11% (2/19) for patients with NC, other solid tumours and DLBCL, respectively. Responding tumours had evidence of deregulated MYC expression.

Conclusions: This trial establishes the safety, favourable pharmacokinetics, evidence of target engagement and preliminary single-agent activity of RO6870810. Responses in patients with NC, other solid tumours and DLBCL provide proof-of-principle for BET inhibition in MYC-driven cancers. The results support further exploration of RO6870810 as monotherapy and in combinations.

Clinical trials registration: NCT01987362.

Conflict of interest statement

All authors have received grants and non-financial or other support from F. Hoffmann-La Roche during the conduct of the study. Editorial support, funded by the sponsor, was provided by an independent medical writer under the guidance of the authors. G.I.S. has served on advisory boards for Roche, Lilly, Pfizer, Bicycle Pharmaceuticals, Merck/EMD Serono, Sierra Oncology, G1 Therapeutics, Fusion Pharmaceuticals, Cybrexa Therapeutics, Bayer, Ipsen, Astex and Almac and has received sponsored research funding from Lilly, Pfizer, Array BioPharma, Merck, Merck/EMD Serono and Sierra Oncology. P.L. has participated in data safety monitoring boards from Chiltern, Agios, Five Prime, Parexel, Halozyme, Novella Clinical and Tyme and in advisory boards from Alexion, Ariad, CytomX, Roche/Genentech, GenMab, Glenmark, Ignyta, Menarini, Novartis, Omniox and Takeda. K.T.D. has participated in advisory boards for QED Therapeutics and Seattle Genetics. C.A.J. has received honorarium or consulting fees from Kite Pharma, Precision Biosciences, Pfizer, Bayer, Novartis, Celgene and Humanigen. U.V. has received research grants from Bristol Myers Squibb, Astellas and Pfizer and personal fees from Bayer, Bristol Myers Squibb, Astellas and Pfizer. A.W. has participated in a speaker bureau and advisory board for Array BioPharma. P.F.C. has received research funding from Roche/Genentech and ADC Therapeutics and participated in speaker bureaus from Celgene and advisory boards for Kite Pharma, Genentech, and Fate Therapeutics. C.A.F. has received research grants from National Institutes of Health, GlaxoSmithKline, C4 Therapeutics, Curis and Constellation and personal fees from GlaxoSmithKline. E.L.-T. and M.D. were employees of Roche at the time of the study and have Roche stock. F.B., W.E.P., J.Z., and S.P. were employees of Roche at the time of the study. M.K. is an employee of Roche and has Roche stock. P.A. has consulted for Merck, Bristol Myers Squibb, Pfizer, Affimed, Adaptive, Infinity, ADC Therapeutics, Celgene, Morphosys, Daiichi Sankyo, Miltenyi, and Tessa and received institutional research funding from Merck, Bristol Myers Squibb, Affimed, Adaptive, Roche, Tensha, Otsuka, Sigma Tau, Genentech and IGM. The following authors have nothing additional to disclose beyond the support received for this work: A.D. and J.P.E.

Figures

Fig. 1. RO6870810 pharmacokinetic profile.
Fig. 1. RO6870810 pharmacokinetic profile.
a Plasma concentration-time profile on days 1 and 15 of cycle 1. b Dose-exposure relationship (cycle 1, day 1).
Fig. 2. RO6870810 pharmacodynamic profiling as evidence…
Fig. 2. RO6870810 pharmacodynamic profiling as evidence of target engagement.
a Mean (± SD) change in CD11b by RO6870810 dose on days 1 and 15 of cycle 1 and day 1 of cycle 2. b Median of mean change in CD11b by RO6870810 dose (cycle 1, day 15). Results are based on the average of three measurements per patient. c Change in CD11b by RO6870810 concentration and LOESS curve. Results are based on three observations per patient on cycle 1, day 15.
Fig. 3. Changes from baseline to best…
Fig. 3. Changes from baseline to best response in measurements of target lesions.
Percentage change from baseline to best response in target lesions is shown in patients with solid tumours (a), NC (b) and diffuse large B-cell lymphoma (DLBCL) (c). Only the largest decrease in the sum of the product or longest diameters for each eligible patient is shown in each figure. For patients with solid tumours, those with missing (n = 5) responses were excluded; one patient with prostate cancer who achieved SD as best response did not have a tumour diameter measurement and is not shown. Six patients with DLBCL without postbaseline tumour assessment were excluded. Among patients with solid tumours, a PR was reported in a patient with salivary gland cancer; SD was reported in patients with salivary gland cancer (n = 4), colorectal cancer (n = 5), oesophageal carcinoma (n = 1), non-small cell lung cancer (n = 1), prostate cancer (n = 7), breast cancer (n = 2), pancreatic cancer (n = 1), uterine cancer (n = 1), ovarian cancer (n = 2) and hepatic cancer (n = 1); PD was reported in patients with squamous cell carcinoma (n = 1), colorectal cancer (n = 4), non-small cell lung cancer (n = 1), poorly differentiated carcinoma of unknown origin (n = 1), pancreatic cancer (n = 1), breast cancer (n = 2), ovarian cancer (n = 1), gastrointestinal stromal tumour (n = 1), fallopian tube cancer (n = 1), endometrial cancer (n = 1) and adenocarcinoma pancreas (n = 1); and a patient with colorectal cancer had an unknown response.

References

    1. Kouzarides T. Chromatin modifications and their function. Cell. 2007;128:693–705.
    1. Drazic A, Myklebust LM, Ree R, Arnesen T. The world of protein acetylation. Biochim Biophys. Acta. 2016;1864:1372–1401.
    1. Zeng L, Zhou MM. Bromodomain: an acetyl-lysine binding domain. FEBS Lett. 2002;513:124–128.
    1. Perez-Salvia M, Esteller M. Bromodomain inhibitors and cancer therapy: from structures to applications. Epigenetics. 2017;12:323–339.
    1. Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov O, et al. Selective inhibition of BET bromodomains. Nature. 2010;468:1067–1073.
    1. Wang CY, Filippakopoulos P. Beating the odds: BETs in disease. Trends Biochem. Sci. 2015;40:468–479.
    1. Sahai V, Redig AJ, Collier KA, Eckerdt FD, Munshi HG. Targeting BET bromodomain proteins in solid tumors. Oncotarget. 2016;7:53997–54009.
    1. Ramadoss M, Mahadevan V. Targeting the cancer epigenome: synergistic therapy with bromodomain inhibitors. Drug Discov. Today. 2018;23:76–89.
    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;146:904–917.
    1. Leal AS, Williams CR, Royce DB, Pioli PA, Sporn MB, Liby KT. Bromodomain inhibitors, JQ1 and I-BET 762, as potential therapies for pancreatic cancer. Cancer Lett. 2017;394:76–87.
    1. Ceribelli M, Kelly PN, Shaffer AL, Wright GW, Xiao W, Yang Y, et al. Blockade of oncogenic IkappaB kinase activity in diffuse large B-cell lymphoma by bromodomain and extraterminal domain protein inhibitors. Proc. Natl Acad. Sci. USA. 2014;111:11365–11370.
    1. Zhu H, Bengsch F, Svoronos N, Rutkowski MR, Bitler BG, Allegrezza MJ, et al. BET bromodomain inhibition promotes anti-tumor immunity by suppressing PD-L1 expression. Cell Rep. 2016;16:2829–2837.
    1. Hogg SJ, Vervoort SJ, Deswal S, Ott CJ, Li J, Cluse LA, et al. BET-bromodomain inhibitors engage the host immune system and regulate expression of the immune checkpoint ligand PD-L1. Cell Rep. 2017;18:2162–2174.
    1. Cioffi M, Trabulo SM, Vallespinos M, Raj D, Kheir TB, Lin ML, et al. The miR-25-93-106b cluster regulates tumor metastasis and immune evasion via modulation of CXCL12 and PD-L1. Oncotarget. 2017;8:21609–21625.
    1. Chapuy B, McKeown MR, Lin CY, Monti S, Roemer MG, Qi J, et al. Discovery and characterization of super-enhancer-associated dependencies in diffuse large B cell lymphoma. Cancer Cell. 2013;24:777–790.
    1. French CA, Ramirez CL, Kolmakova J, Hickman TT, Cameron MJ, Thyne ME, 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;27:2237–2242.
    1. French CA, Rahman S, Walsh EM, Kuhnle S, Grayson AR, Lemieux ME, et al. NSD3-NUT fusion oncoprotein in NUT midline carcinoma: implications for a novel oncogenic mechanism. Cancer Discov. 2014;4:928–941.
    1. Grayson AR, Walsh EM, Cameron MJ, Godec J, Ashworth T, Ambrose JM, et al. MYC, a downstream target of BRD-NUT, is necessary and sufficient for the blockade of differentiation in NUT midline carcinoma. Oncogene. 2014;33:1736–1742.
    1. Boi M, Gaudio E, Bonetti P, Kwee I, Bernasconi E, Tarantelli C, et al. The BET bromodomain inhibitor OTX015 affects pathogenetic pathways in preclinical B-cell tumor models and synergizes with targeted drugs. Clin. Cancer Res. 2015;21:1628–1638.
    1. Trabucco SE, Gerstein RM, Evens AM, Bradner JE, Shultz LD, Greiner DL, et al. Inhibition of bromodomain proteins for the treatment of human diffuse large B-cell lymphoma. Clin. Cancer Res. 2015;21:113–122.
    1. Savage KJ, Johnson NA, Ben-Neriah S, Connors JM, Sehn LH, Farinha P, et al. MYC gene rearrangements are associated with a poor prognosis in diffuse large B-cell lymphoma patients treated with R-CHOP chemotherapy. Blood. 2009;114:3533–3537.
    1. Green TM, Young KH, Visco C, Xu-Monette ZY, Orazi A, Go RS, et al. Immunohistochemical double-hit score is a strong predictor of outcome in patients with diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J. Clin. Oncol. 2012;30:3460–3467.
    1. Mensah AA, Cascione L, Gaudio E, Tarantelli C, Bomben R, Bernasconi E, et al. Bromodomain and extra-terminal domain inhibition modulates the expression of pathologically relevant microRNAs in diffuse large B-cell lymphoma. Haematologica. 2018;103:2049–2058.
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur. J. Cancer. 2009;45:228–247.
    1. Cheson BD, Fisher RI, Barrington SF, Cavalli F, Schwartz LH, Zucca E, et al. Recommendations for initial evaluation, staging, and response assessment of hodgkin and non-hodgkin lymphoma: the lugano classification. J. Clin. Oncol. 2014;32:3059–3068.
    1. Stathis A, Bertoni F. BET proteins as targets for anticancer treatment. Cancer Discov. 2018;8:24–36.
    1. Doroshow DB, Eder JP, LoRusso PM. BET inhibitors: a novel epigenetic approach. Ann. Oncol. 2017;28:1776–1787.
    1. Amorim S, Stathis A, Gleeson M, Iyengar S, Magarotto V, Leleu X, 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:e196–e204.
    1. Berthon C, Raffoux E, Thomas X, Vey N, Gomez-Roca C, Yee K, et al. Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study. Lancet Haematol. 2016;3:e186–e195.
    1. Blum KA, Abramson J, Maris M, Flinn I, Goy A, Mertz J, et al. A phase I study of CPI-0610, a bromodomain and extra terminal protein (BET) inhibitor in patients with relapsed or refractory lymphoma. Ann. Oncol. 2018;29:iii7–iii9.
    1. Lewin J, Soria JC, Stathis A, Delord JP, Peters S, Awada 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;36:3007–3014.
    1. Piha-Paul, S. A., Hann, C. L., French, C. A., Cousin, S., Brana, I., Cassier, P. A. et al. Phase 1 study of molibresib (GSK525762), a bromodomain and extra-terminal domain protein inhibitor, in NUT carcinoma and other solid tumors. JNCI Cancer Spectrum4, pkz093 (2019).
    1. Aftimos PG, Bechter O, Awada A, Jungels C, Dumez H, Huyvaert N, et al. Phase I first-in-man trial of a novel bromodomain and extra-terminal domain (BET) inhibitor (BI 894999) in patients (pts) with advanced solid tumors. J. Clin. Oncol. 2018;36:(abstract 2504).
    1. Hilton J, Cristea MC, Voskoboynik M, Postel-Vinay S, Edenfield W, Gavai A, et al. 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:abstr. 4110.
    1. Antolic A, Wakimoto H, Jiao Z, Gorham JM, DePalma SR, Lemieux ME, et al. BET bromodomain proteins regulate transcriptional reprogramming in genetic dilated cardiomyopathy. JCI Insight. 2020;5:138687.
    1. Piquereau J, Boet A, Péchoux C, Antigny F, Lambert M, Gressette M, et al. The BET bromodomain inhibitor I-BET-151 induces structural and functional alterations of the heart mitochondria in healthy male mice and rats. Int. J. Mol. Sci. 2019;20:1527.
    1. Chau NG, Ma C, Danga K, Al-Sayegh H, Sridharan M, Nardi V, et al. A novel prognostic risk classification model for NUT midline carcinoma: a largest cohort analysis from the NMC registry. J. Clin. Oncol. 2018;36:abstract 6085.
    1. Petrich AM, Gandhi M, Jovanovic B, Castillo JJ, Rajguru S, Yang DT, et al. Impact of induction regimen and stem cell transplantation on outcomes in double-hit lymphoma: a multicenter retrospective analysis. Blood. 2014;124:2354–2361.
    1. Johnson NA, Slack GW, Savage KJ, Connors JM, Ben-Neriah S, Rogic S, et al. Concurrent expression of MYC and BCL2 in diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J. Clin. Oncol. 2012;30:3452–2459.
    1. Chapuy B, Stewart C, Dunford AJ, Kim J, Kamburov A, Redd RA, et al. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat. Med. 2018;24:679–690.
    1. Fong CY, Gilan O, Lam EY, Rubin AF, Ftouni S, Tyler D, et al. BET inhibitor resistance emerges from leukaemia stem cells. Nature. 2015;525:538–542.
    1. Rathert P, Roth M, Neumann T, Muerdter F, Roe JS, Muhar M, et al. Transcriptional plasticity promotes primary and acquired resistance to BET inhibition. Nature. 2015;525:543–547.
    1. Kumar K, Raza SS, Knab LM, Chow CR, Kwok B, Bentrem DJ, et al. GLI2-dependent c-MYC upregulation mediates resistance of pancreatic cancer cells to the BET bromodomain inhibitor JQ1. Sci. Rep. 2015;5:9489.
    1. Shu S, Lin CY, He HH, Witwicki RM, Tabassum DP, Roberts JM, et al. Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer. Nature. 2016;529:413–417.
    1. Kurimchak AM, Shelton C, Duncan KE, Johnson KJ, Brown J, O’Brien S, et al. Resistance to BET bromodomain inhibitors is mediated by kinome reprogramming in ovarian cancer. Cell Rep. 2016;16:1273–1286.
    1. Ma Y, Wang L, Neitzel LR, Loganathan SN, Tang N, Qin L, et al. The MAPK pathway regulates intrinsic resistance to BET inhibitors in colorectal cancer. Clin. Cancer Res. 2017;23:2027–2037.
    1. Jang JE, Eom JI, Jeung HK, Cheong JW, Lee JY, Kim JS, et al. AMPK-ULK1-mediated autophagy confers resistance to BET inhibitor JQ1 in acute myeloid leukemia stem cells. Clin. Cancer Res. 2017;23:2781–2794.
    1. Zhao Y, Liu Q, Acharya P, Stengel KR, Sheng Q, Zhou X, et al. High-resolution mapping of RNA polymerases identifies mechanisms of sensitivity and resistance to BET inhibitors in t(8;21) AML. Cell Rep. 2016;16:2003–2016.
    1. Hogg SJ, Newbold A, Vervoort SJ, Cluse LA, Martin BP, Gregory GP, et al. BET inhibition induces apoptosis in aggressive B-cell lymphoma via epigenetic regulation of BCL-2 family members. Mol. Cancer Ther. 2016;15:2030–2041.

Source: PubMed

3
Subskrybuj