Identification of a genetic signature enriching for response to ibrutinib in relapsed/refractory follicular lymphoma in the DAWN phase 2 trial

Sriram Balasubramanian, Brendan Hodkinson, Stephen J Schuster, Nathan H Fowler, Judith Trotman, Georg Hess, Bruce D Cheson, Michael Schaffer, Steven Sun, Sanjay Deshpande, Jessica Vermeulen, Gilles Salles, Ajay K Gopal, Sriram Balasubramanian, Brendan Hodkinson, Stephen J Schuster, Nathan H Fowler, Judith Trotman, Georg Hess, Bruce D Cheson, Michael Schaffer, Steven Sun, Sanjay Deshpande, Jessica Vermeulen, Gilles Salles, Ajay K Gopal

Abstract

Background: The single-arm DAWN trial (NCT01779791) of ibrutinib monotherapy in patients with relapsed/refractory follicular lymphoma (FL) showed an overall response rate (ORR) of 20.9% and a median response duration of 19.4 months. This biomarker analysis of the DAWN dataset sought to determine genetic classifiers for prediction of response to ibrutinib treatment.

Methods: Whole exome sequencing was performed on baseline tumor samples. Potential germline variants were excluded; a custom set of 1216 cancer-related genes was examined. Responder- versus nonresponder-associated variants were identified using Fisher's exact test. Classifiers with increasing numbers of genes were created using a greedy algorithm that repeatedly selected genes, adding the most nonresponders to the existing "predicted nonresponders" set and were evaluated with 10-fold cross-validation.

Results: Exome data were generated from 88 patient samples and 13,554 somatic mutation variants were inferred. Response data were available for 83 patients (17 responders, 66 nonresponders). Each sample showed 100 to >500 mutated genes, with greater variance across nonresponders. The overall variant pattern was consistent with previous FL studies; 75 genes had mutations in >10% of patients, including genes previously reported as associated with FL. Univariate analysis yielded responder-associated genes FANCA, HISTH1B, ANXA6, BTG1, and PARP10, highlighting the importance of functions outside of B-cell receptor signaling, including epigenetic processes, DNA damage repair, cell cycle/proliferation, and cell motility/invasiveness. While nonresponder-associated genes included well-known TP53 and CARD11, genetic classifiers developed using nonresponder-associated genes included ATP6AP1, EP400, ARID1A, SOCS1, and TBL1XR1, suggesting resistance to ibrutinib may be related to broad biological functions connected to epigenetic modification, telomere maintenance, and cancer-associated signaling pathways (mTOR, JAK/STAT, NF-κB).

Conclusion: The results from univariate and genetic classifier analyses provide insights into genes associated with response or resistance to ibrutinib in FL and identify a classifier developed using nonresponder-associated genes, which warrants further investigation.

Trial registration: NCT01779791.

Keywords: biomarkers; genetic variants; lymphoma; mutations.

Conflict of interest statement

SB: Employment: Janssen Research & Development; Equity ownership: Janssen Research & Development. BH, MS, SS, SD, and JV: Employment: Janssen Research & Development. SJS: Consultancy and research funding: Novartis, Pharmacyclics, Celgene; Research funding: Gilead, Janssen Research & Development, Hoffmann‐La Roche, Merck; Board of directors or advisory committees: Nordic Nanovector; Consultancy: Genentech, Acerta. NHF: Consulting or advisory role: Pharmacyclics, Janssen; Research funding: Pharmacyclics, Janssen. JT: Research funding to institution: Janssen, PCYC, Roche, BeiGene, Takeda, Celgene; Cooperative group funding: Janssen; Advisory role (unremunerated): Janssen, Roche, Celgene, Takeda. GH: Research support: Roche, Pfizer, Celgene, CTI; Consultant: Roche, Pfizer, Janssen, Celgene, AbbVie; Honoraria: Roche, Pfizer, Janssen, Celgene, AbbVie; Advisory board: Roche, Pfizer, Janssen, Celgene, AbbVie. BDC: Advisory board: Acerta, AbbVie, Roche‐Genentech, Celgene, TG Therapeutics; Research funding to institution: Gilead, Pharmacyclics, Acerta, TG Therapeutics, AbbVie. GS: Research funding: Roche, Celgene; Consultancy: Novartis; Honoraria: AbbVie, Acerta, Amgen, Celgene, Epizyme, Gilead, Janssen, Merck, Morphosys, Novartis, Pfizer, Roche, Servier, Takeda, Celgene, Mundipharma. AKG: Research funding: Merck, Janssen, Spectrum, Takeda, Bristol Myers Squibb, Pfizer, Seattle Genetics, Gilead; Donations: Frank and Betty Vandermeer; Consulting fees: Seattle Genetics, Gilead, Janssen, Brim, Aptevo, Genzyme.

© 2021 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Cancer‐associated genes of interest that are mutated in more than three patients with follicular lymphoma. Left panel shows the percentage of individuals with a mutation in each gene; right panel shows the distribution of mutations in those genes in the 83 patients with response/nonresponse data
FIGURE 2
FIGURE 2
Heatmap of ranked nonresponder gene mutations in ibrutinib‐treated patients with follicular lymphoma
FIGURE 3
FIGURE 3
Mean overall response rate (ORR) of predicted responders (gray line) based on 10‐fold cross‐validation for different responder/nonresponder classification models containing an increasing number of genes. The dotted blue line represents the ORR of the entire patient cohort regardless of classification. ORR, overall response rate
FIGURE 4
FIGURE 4
Plots of somatic mutations in the genes of interest associated with the lack of response to ibrutinib: (A) ATP6AP1, (B) EP400, (C) ARID1A, (D) SOCS1, (E) TBL1XR1, and (F) CARD 11. DEL = the mutation was predicted to be deleterious by metaSVM

References

    1. Dreyling M, Ghielmini M, Rule S, Salles G, Vitolo U, Ladetto M. Newly diagnosed and relapsed follicular lymphoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow‐up. Ann Oncol. 2017;28(12):3109.
    1. Teras LR, DeSantis CE, Cerhan JR, Morton LM, Jemal A, Flowers CR. 2016 US lymphoid malignancy statistics by World Health Organization subtypes. CA Cancer J Clin. 2016;66(6):443‐459.
    1. National Comprehensive Cancer Network . NCCN Clinical Practice Guidelines in Oncology. B‐cell lymphomas. Version 4. 2015. Available from:
    1. Casulo C, Nastoupil L, Fowler NH, Friedberg JW, Flowers CR. Unmet needs in the first‐line treatment of follicular lymphoma. Ann Oncol. 2017;28(9):2094‐2106.
    1. Bai B, Huang HQ. Individualized management of follicular lymphoma. Chin Clin Oncol. 2015;4(1):7.
    1. Cheah CY, Fowler NH, Wang ML. Breakthrough therapies in B‐cell non‐Hodgkin lymphoma. Ann Oncol. 2016;27(5):778‐787.
    1. Gayko U, Fung M, Clow F, et al. Development of the Bruton's tyrosine kinase inhibitor ibrutinib for B cell malignancies. Ann N Y Acad Sci. 2015;1358:82‐94.
    1. IMBRUVICA® (ibrutinib) [prescribing information] . Pharmacyclics LLC, Sunnyvale, CA, USA, Janssen Biotech, Inc., Horsham, PA, USA; 2018.
    1. Burger JA, Tedeschi A, Barr PM, et al. Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med. 2015;373(25):2425‐2437.
    1. Byrd JC, Brown JR, O'Brien S, et al. Ibrutinib versus ofatumumab in previously treated chronic lymphoid leukemia. N Engl J Med. 2014;371(3):213‐223.
    1. Byrd JC, Furman RR, Coutre SE, et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med. 2013;369(1):32‐42.
    1. Chanan‐Khan A, Cramer P, Demirkan F, et al. Ibrutinib combined with bendamustine and rituximab compared with placebo, bendamustine, and rituximab for previously treated chronic lymphocytic leukaemia or small lymphocytic lymphoma (HELIOS): a randomised, double‐blind, phase 3 study. Lancet Oncol. 2016;17(2):200‐211.
    1. Dimopoulos MA, Trotman J, Tedeschi A, et al. Ibrutinib for patients with rituximab‐refractory Waldenstrom's macroglobulinaemia (iNNOVATE): an open‐label substudy of an international, multicentre, phase 3 trial. Lancet Oncol. 2017;18(2):241‐250.
    1. Wang ML, Rule S, Martin P, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle‐cell lymphoma. N Engl J Med. 2013;369(6):507‐516.
    1. IMBRUVICA (ibrutinib) [summary of product characteristics] . Janssen Pharmaceutica NV, Beerse, Belgium; 2018.
    1. Advani RH, Buggy JJ, Sharman JP, et al. Bruton tyrosine kinase inhibitor ibrutinib (PCI‐32765) has significant activity in patients with relapsed/refractory B‐cell malignancies. J Clin Oncol. 2013;31(1):88‐94.
    1. Bartlett NL, Costello BA, LaPlant BR, et al. Single‐agent ibrutinib in relapsed or refractory follicular lymphoma: a phase 2 consortium trial. Blood. 2018;131(2):182‐190.
    1. Fowler N, Boyd TE, Sharman JP, et al. Long‐term follow‐up and analysis of dose groups with ibrutinib in relapsed follicular lymphoma [abstract]. Presented at: American Society of Hematology Annual Meeting; 5‐8 December 2015; Orlando, FL. 2015. Abstract 2706.
    1. Gopal AK, Schuster SJ, Fowler NH, et al. Ibrutinib as treatment for patients with relapsed/refractory follicular lymphoma: results from the open‐label, multicenter, phase II DAWN study. J Clin Oncol. 2018;36(23):2405‐2412.
    1. Krysiak K, Gomez F, White BS, et al. Recurrent somatic mutations affecting B‐cell receptor signaling pathway genes in follicular lymphoma. Blood. 2017;129(4):473‐483.
    1. Taylor J, Xiao W, Abdel‐Wahab O. Diagnosis and classification of hematologic malignancies on the basis of genetics. Blood. 2017;130(4):410‐423.
    1. Cheson BD, Pfistner B, Juweid ME, et al. Revised response criteria for malignant lymphoma. J Clin Oncol. 2007;25(5):579‐586.
    1. R Core Team . R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. 2014. Available from:
    1. Bösl MW, Osterode E, Bararia D, et al. STAT6 is recurrently and significantly mutated in follicular lymphoma and enhances the IL‐4 induced expression of membrane‐bound and soluble CD23 [abstract]. Presented at: American Society of Hematology Annual Meeting; 5‐8 December 2015; Orlando, FL. 2015. Abstract 126.
    1. Morin RD, Johnson NA, Severson TM, et al. Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B‐cell lymphomas of germinal‐center origin. Nat Genet. 2010;42(2):181‐185.
    1. Li H, Kaminski MS, Li Y, et al. Mutations in linker histone genes HIST1H1 B, C, D, and E; OCT2 (POU2F2); IRF8; and ARID1A underlying the pathogenesis of follicular lymphoma. Blood. 2014;123(10):1487‐1498.
    1. Mottok A, Renné C, Seifert M, et al. Inactivating SOCS1 mutations are caused by aberrant somatic hypermutation and restricted to a subset of B‐cell lymphoma entities. Blood. 2009;114(20):4503‐4506.
    1. Wang X, Haswell JR, Roberts CW. Molecular pathways: SWI/SNF (BAF) complexes are frequently mutated in cancer–mechanisms and potential therapeutic insights. Clin Cancer Res. 2014;20(1):21‐27.
    1. Yu M, Schreek S, Cerni C, et al. PARP‐10, a novel Myc‐interacting protein with poly(ADP‐ribose) polymerase activity, inhibits transformation. Oncogene. 2005;24(12):1982‐1993.
    1. Nicolae CM, Aho ER, Vlahos AHS, et al. The ADP‐ribosyltransferase PARP10/ARTD10 interacts with proliferating cell nuclear antigen (PCNA) and is required for DNA damage tolerance. J Biol Chem. 2014;289(19):13627‐13637.
    1. Flores‐Pérez A, Rafaelli LE, Ramírez‐Torres N, et al. RAD50 targeting impairs DNA damage response and sensitizes human breast cancer cells to cisplatin therapy. Cancer Biol Ther. 2014;15(6):777‐788.
    1. Hosoya N, Miyagawa K. Targeting DNA damage response in cancer therapy. Cancer Sci. 2014;105(4):370‐388.
    1. Litman R, Gupta R, Brosh RM Jr, Cantor SB. BRCA‐FA pathway as a target for anti‐tumor drugs. Anticancer Agents Med Chem. 2008;8(4):426‐430.
    1. Zheng H‐C, Li J, Shen D‐F, et al. BTG1 expression correlates with pathogenesis, aggressive behaviors and prognosis of gastric cancer: a potential target for gene therapy. Oncotarget. 2015;6(23):19685‐19705.
    1. Scheijen B, Boer JM, Marke R, et al. Tumor suppressors BTG1 and IKZF1 cooperate during mouse leukemia development and increase relapse risk in B‐cell precursor acute lymphoblastic leukemia patients. Haematologica. 2017;102(3):541‐551.
    1. Koumangoye RB, Nangami GN, Thompson PD, Agboto VK, Ochieng J, Sakwe AM. Reduced annexin A6 expression promotes the degradation of activated epidermal growth factor receptor and sensitizes invasive breast cancer cells to EGFR‐targeted tyrosine kinase inhibitors. Mol Cancer. 2013;12(1):167.
    1. Okosun J, Wolfson RL, Wang J, et al. Recurrent mTORC1‐activating RRAGC mutations in follicular lymphoma. Nat Genet. 2016;48(2):183‐188.
    1. Zhang J, Li H, Yu JP, Wang SE, Ren XB. Role of SOCS1 in tumor progression and therapeutic application. Int J Cancer. 2012;130(9):1971‐1980.
    1. Pastore A, Jurinovic V, Kridel R, et al. Integration of gene mutations in risk prognostication for patients receiving first‐line immunochemotherapy for follicular lymphoma: a retrospective analysis of a prospective clinical trial and validation in a population‐based registry. Lancet Oncol. 2015;16(9):1111‐1122.
    1. Wang J, Liu X, Hong Y, et al. Ibrutinib, a Bruton's tyrosine kinase inhibitor, exhibits antitumoral activity and induces autophagy in glioblastoma. J Exp Clin Cancer Res. 2017;36(1):96.
    1. Smith JA, White EA, Sowa ME, et al. Genome‐wide siRNA screen identifies SMCX, EP400, and Brd4 as E2‐dependent regulators of human papillomavirus oncogene expression. Proc Natl Acad Sci USA. 2010;107(8):3752‐3757.
    1. Rahmanto YS, Jung J‐G, Wu R‐C, et al. Inactivating ARID1A tumor suppressor enhances TERT transcription and maintains telomere length in cancer cells. J Biol Chem. 2016;291(18):9690‐9699.
    1. Guo A, Lu P, Coffey G, Conley P, Pandey A, Wang YL. Dual SYK/JAK inhibition overcomes ibrutinib resistance in chronic lymphocytic leukemia: Cerdulatinib, but not ibrutinib, induces apoptosis of tumor cells protected by the microenvironment. Oncotarget. 2017;8(8):12953‐12967.
    1. Amin AD, Peters TL, Li L, et al. Diffuse large B‐cell lymphoma: can genomics improve treatment options for a curable cancer? Cold Spring Harb Mol Case Stud. 2017;3(3):a001719.
    1. Zhang SQ, Smith SM, Zhang SY, Lynn WY. Mechanisms of ibrutinib resistance in chronic lymphocytic leukaemia and non‐Hodgkin lymphoma. Br J Haematol. 2015;170(4):445‐456.
    1. Chan W, Schaffer TB, Pomerantz JL. A quantitative signaling screen identifies CARD11 mutations in the CARD and LATCH domains that induce Bcl10 ubiquitination and human lymphoma cell survival. Mol Cell Biol. 2013;33(2):429‐443.
    1. Buchbinder DK, Stinson JR, Nugent DJ, et al. A novel gain‐of‐function mutation in the CARD domain of CARD11 (C49Y) results in BENTA disease. Blood. 2013;122(21):3485.
    1. Buchbinder D, Stinson JR, Nugent DJ, et al. Mild B‐cell lymphocytosis in patients with a CARD11 C49Y mutation. J Allergy Clin Immunol. 2015;136(3):819‐821 e811.

Source: PubMed

3
Subscribe