Predicting Toxicity and Response to Pembrolizumab Through Germline Genomic HLA Class 1 Analysis

Marco A J Iafolla, Cindy Yang, Vinod Chandran, Melania Pintilie, Quan Li, Philippe L Bedard, Aaron Hansen, Stephanie Lheureux, Anna Spreafico, Albiruni A Razak, Sevan Hakgor, Amanda Giesler, Trevor J Pugh, Lillian L Siu, Marco A J Iafolla, Cindy Yang, Vinod Chandran, Melania Pintilie, Quan Li, Philippe L Bedard, Aaron Hansen, Stephanie Lheureux, Anna Spreafico, Albiruni A Razak, Sevan Hakgor, Amanda Giesler, Trevor J Pugh, Lillian L Siu

Abstract

Background: Human leukocyte antigen class 1 (HLA-1)-dependent immune activity is linked to autoimmune diseases. HLA-1-dependent CD8+ T cells are required for immune checkpoint blockade antitumor activity. It is unknown if HLA-1 genotype is predictive of toxicity to immune checkpoint blockade.

Methods: Patients with advanced solid tumors stratified into 5 cohorts received single agent pembrolizumab (anti-programmed cell death-1) 200 mg intravenously every 3 weeks in an investigator-initiated phase II trial (Investigator-Initiated Phase II Study of Pembrolizumab Immunological Response Evaluation study, NCT02644369). Germline whole-exome sequencing of peripheral blood mononuclear cells was performed using the Illumina HiSeq2500 platform. HLA-1 haplotypes were predicted from whole-exome sequencing using HLAminer and HLAVBSeq. Heterozygosity of HLA-A, -B, and -C, individual HLA-1 alleles, and HLA haplotype dimorphism at positions -21 M and -21 T of the HLA-A and -B leader sequence were analyzed as predictors of toxicity defined as grade 2 or greater immune-related adverse events and clinical benefit defined as complete or partial response, or stable disease for 6 or more cycles of pembrolizumab. Statistical significance tests were 2-sided.

Results: In the overall cohort of 101 patients, the frequency of toxicity and clinical benefit from pembrolizumab was 22.8% and 25.7%, respectively. There was no association between any of the HLA-1 loci or alleles with toxicity. HLA-C heterozygosity had an association with decreased clinical benefit relative to HLA-C homozygosity when controlling for cohort (odds ratio = 0.28, 95% confidence interval = 0.09 to 0.91, P = .04). HLA-A and -B haplotype -21 M/T dimorphism and heterozygosity of HLA-A, -B, and -C were not predictive of outcomes.

Conclusions: HLA-C heterozygosity may predict decreased response to pembrolizumab. Prospective validation is required.

© The Author(s) 2020. Published by Oxford University Press.

Figures

Figure 1.
Figure 1.
Consolidated Standards of Reporting Trials (CONSORT) diagram of patient allocation and outcomes. CONSORT diagram displaying all relevant details regarding the 101 patients used in this human leukocyte antigen class 1 (HLA-1) analysis from the Investigator-Initiated Phase II Study of Pembrolizumab Immunological Response Evaluation (INSPIRE) study. Note that patients can develop more than 1 type of toxicity event. CB = clinical benefit; CR = complete response; HGSOC = high-grade serous ovarian carcinoma; HNSCC = head and neck squamous cell carcinoma; NE = nonevaluable; PD = progressive disease; PR = partial response; SD = stable disease; TNBC = triple-negative breast cancer.
Figure 2.
Figure 2.
Survival and progression outcomes. Kaplan-Meier curves of (A) overall survival (OS) and (B) progression-free survival (PFS) after dichotomizing patients with germline heterozygosity of human leukocyte antigen (HLA)-A, -B, and -C compared with homozygosity of at least 1 HLA loci. P values are adjusted for cohort and calculated using the Wald test. CI = confidence interval; HR = hazard ratio.

References

    1. Farkona S, Diamandis EP, Blasutig IM.. Cancer immunotherapy: the beginning of the end of cancer? BMC Med. 2016;14(1):73.
    1. Cully M. Combinations with checkpoint inhibitors at wavefront of cancer immunotherapy. Nat Rev Drug Discov. 2015;14(6):374–375.
    1. Sunshine J, Taube JM.. PD-1/PD-L1 inhibitors. Curr Opin Pharmacol. 2015;23:32–38.
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, et al.Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373(1):23–34.
    1. Villadolid J, Amin A.. Immune checkpoint inhibitors in clinical practice: update on management of immune-related toxicities. Transl Lung Cancer Res. 2015;4(5):560–575.
    1. Weber JS, D'Angelo SP, Minor D, et al.Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015;16(4):375–384.
    1. Robert C, Schachter J, Long GV, et al.Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015;372(26):2521–2532.
    1. Brahmer J, Reckamp KL, Baas P, et al.Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015;373(2):123–135.
    1. Gibney GT, Weiner LM, Atkins MB.. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016;17(12):e542–e551.
    1. Menzies AM, Johnson DB, Ramanujam S, et al.Anti-PD-1 therapy in patients with advanced melanoma and preexisting autoimmune disorders or major toxicity with ipilimumab. Ann Oncol. 2017;28(2):368–376.
    1. Doherty PC, Zinkernagel RM.. A biological role for the major histocompatibility antigens. Lancet. 1975;1(7922):1406–1409.
    1. Brooks AG, Boyington JC, Sun PD.. Natural killer cell recognition of HLA class I molecules. Rev Immunogenet. 2000;2(3):433–448.
    1. Aptsiauri N, Cabrera T, Garcia-Lora A, Lopez-Nevot MA, Ruiz-Cabello F, Garrido F.. MHC class I antigens and immune surveillance in transformed cells. Int Rev Cytol. 2007;256:139–189.
    1. Gubin MM, Zhang X, Schuster H, et al.Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature. 2014;515(7528):577–581.
    1. Tran E, Ahmadzadeh M, Lu YC, et al.Immunogenicity of somatic mutations in human gastrointestinal cancers. Science. 2015;350(6266):1387–1390.
    1. Tran E, Robbins PF, Lu YC, et al.T-cell transfer therapy targeting mutant KRAS in cancer. N Engl J Med. 2016;375(23):2255–2262.
    1. Beldi-Ferchiou A, Caillat-Zucman S.. Control of NK cell activation by immune checkpoint molecules. Int J Mol Sci. 2017;18(10):2129.
    1. Benci JL, Johnson LR, Choa R, et al.Opposing functions of interferon coordinate adaptive and innate immune responses to cancer immune checkpoint blockade. Cell. 2019;178(4):933–948.e14.
    1. McGranahan N, Rosenthal R, Hiley CT, et al.Allele-specific HLA loss and immune escape in lung cancer evolution. Cell. 2017;171(6):1259–1271.e11.
    1. Campoli M, Ferrone S.. HLA antigen changes in malignant cells: epigenetic mechanisms and biologic significance. Oncogene. 2008;27(45):5869–5885.
    1. Hiraki A, Fujii N, Murakami T, et al.High frequency of allele-specific down-regulation of HLA class I expression in lung cancer cell lines. Anticancer Res. 2004;24(3a):1525–1528.
    1. Mehta AM, Jordanova ES, Kenter GG, Ferrone S, Fleuren GJ.. Association of antigen processing machinery and HLA class I defects with clinicopathological outcome in cervical carcinoma. Cancer Immunol Immunother. 2007;57(2):197–206.
    1. Terasaki PI, Tiwari JL.. HLA and Disease Associations. New York: Springer-Verlag; 1985.
    1. Eagar TN, Karandikar NJ, Bluestone JA, Miller SD.. The role of CTLA-4 in induction and maintenance of peripheral T cell tolerance. Eur J Immunol. 2002;32(4):972–981.
    1. Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ.. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol. 2007;8(3):239–245.
    1. Horowitz A, Djaoud Z, Nemat-Gorgani N, et al.Class I HLA haplotypes form two schools that educate NK cells in different ways. Sci Immunol. 2016;1(3):eaag1672.
    1. Common Terminology Criteria for Adverse Events (CTCAE) v4.03. 2010. National Cancer Institute. . Accessed January 5, 2021.
    1. Champiat S, Lambotte O, Barreau E, et al.Management of immune checkpoint blockade dysimmune toxicities: a collaborative position paper. Ann Oncol. 2016;27(4):559–574.
    1. Clouthier DL, Lien SC, Yang SYC, et interim report on the investigator-initiated phase 2 study of pembrolizumab immunological response evaluation (INSPIRE). J Immunother Cancer. 2019;7(1):72.
    1. Chowell D, Morris LGT, Grigg CM, et al.Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science. 2018;359(6375):582–587.
    1. Shukla SA, Rooney MS, Rajasagi M, et al.Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat Biotechnol. 2015;33(11):1152–1158.
    1. Castro A, Ozturk K, Pyke RM, Xian S, Zanetti M, Carter H.. Elevated neoantigen levels in tumors with somatic mutations in the HLA-A, HLA-B, HLA-C and B2M genes. BMC Med Genomics. 2019;12(S6):107.
    1. del Campo AB, Kyte JA, Carretero J, et al.Immune escape of cancer cells with beta2-microglobulin loss over the course of metastatic melanoma. Int J Cancer. 2014;134(1):102–113.
    1. Yoshihama S, Roszik J, Downs I, et al.NLRC5/MHC class I transactivator is a target for immune evasion in cancer. Proc Natl Acad Sci USA. 2016;113(21):5999–6004.
    1. Hugo W, Zaretsky JM, Sun L, et al.Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell. 2016;165(1):35–44.
    1. Le DT, Uram JN, Wang H, et al.PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372(26):2509–2520.
    1. Rizvi NA, Hellmann MD, Snyder A, et al.Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348(6230):124–128.
    1. Riaz N, Havel JJ, Makarov V, et al.Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell. 2017;171(4):934–949.e16.
    1. Seliger B, Kloor M, Ferrone S.. HLA class II antigen-processing pathway in tumors: molecular defects and clinical relevance. Oncoimmunology. 2017;6(2):e1171447.
    1. Johnson DB, Estrada MV, Salgado R, et al.Melanoma-specific MHC-II expression represents a tumour-autonomous phenotype and predicts response to anti-PD-1/PD-L1 therapy. Nat Commun. 2016;7(1):10582.
    1. Roemer MGM, Redd RA, Cader FZ, et al.Major histocompatibility complex class II and programmed death ligand 1 expression predict outcome after programmed death 1 blockade in classic Hodgkin lymphoma. J Clin Oncol. 2018;36(10):942–950.
    1. Rodig SJ, Gusenleitner D, Jackson DG, et al.MHC proteins confer differential sensitivity to CTLA-4 and PD-1 blockade in untreated metastatic melanoma. Sci Transl Med. 2018;10(450):eaar3342.
    1. Johnson DB, Nixon MJ, Wang Y, et al.Tumor-specific MHC-II expression drives a unique pattern of resistance to immunotherapy via LAG-3/FCRL6 engagement. JCI Insight. 2018;3(24):e120360.
    1. Raulet DH, Guerra N.. Oncogenic stress sensed by the immune system: role of natural killer cell receptors. Nat Rev Immunol. 2009;9(8):568–580.
    1. Cerwenka A, Lanier LL.. Natural killer cells, viruses and cancer. Nat Rev Immunol. 2001;1(1):41–49.
    1. Pesce S, Greppi M, Tabellini G, et al.Identification of a subset of human natural killer cells expressing high levels of programmed death 1: a phenotypic and functional characterization. J Allergy Clin Immunol. 2017;139(1):335–346.e3.
    1. Beldi-Ferchiou A, Lambert M, Dogniaux S, et al.PD-1 mediates functional exhaustion of activated NK cells in patients with Kaposi sarcoma. Oncotarget. 2016;7(45):72961–72977.
    1. Segal NH, Naidoo J, Curigliano G, et al.First-in-human dose escalation of monalizumab plus durvalumab, with expansion in patients with metastatic microsatellite-stable colorectal cancer. J Clin Oncol. 2018;36(15_suppl):3540–3541.
    1. Goyette P, Boucher G, Mallon D, et al.; International Inflammatory Bowel Disease Genetics Consortium. High-density mapping of the MHC identifies a shared role for HLA-DRB101:03 in inflammatory bowel diseases and heterozygous advantage in ulcerative colitis. Nat Genet. 2015;47(2):172–179.
    1. Hasan Ali O, Berner F, Bomze D, et al.Human leukocyte antigen variation is associated with adverse events of checkpoint inhibitors. Eur J Cancer. 2019;107:8–14.
    1. Weber JS, Hodi FS, Wolchok JD, et al.Safety profile of nivolumab monotherapy: a pooled analysis of patients with advanced melanoma. J Clin Oncol. 2017;35(7):785–792.
    1. Haratani K, Hayashi H, Chiba Y, et al.Association of immune-related adverse events with nivolumab efficacy in non-small-cell lung cancer. JAMA Oncol. 2018;4(3):374–378.
    1. Propert KJ, Anderson JR.. Assessing the effect of toxicity on prognosis: methods of analysis and interpretation. J Clin Oncol. 1988;6(5):868–870.
    1. Anderson JR, Cain KC, Gelber RD.. Analysis of survival by tumor response and other comparisons of time-to-event by outcome variables. J Clin Oncol. 2008;26(24):3913–3915.

Source: PubMed

3
購読する