Afatinib and Pembrolizumab for Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma (ALPHA Study): A Phase II Study with Biomarker Analysis

Hsiang-Fong Kao, Bin-Chi Liao, Yen-Lin Huang, Huai-Cheng Huang, Chun-Nan Chen, Tseng-Cheng Chen, Yuan-Jing Hong, Ching-Yi Chan, Jean-San Chia, Ruey-Long Hong, Hsiang-Fong Kao, Bin-Chi Liao, Yen-Lin Huang, Huai-Cheng Huang, Chun-Nan Chen, Tseng-Cheng Chen, Yuan-Jing Hong, Ching-Yi Chan, Jean-San Chia, Ruey-Long Hong

Abstract

Purpose: EGFR pathway inhibition may promote anti-programmed cell death protein 1 (PD-1) responses in preclinical models, but how EGFR inhibition affects tumor antigen presentation during anti-PD-1 monotherapy in humans remain unknown. We hypothesized that afatinib, an irreversible EGFR tyrosine kinase inhibitor, would improve outcomes in patients treated with pembrolizumab for recurrent or metastatic head and neck squamous cell carcinoma (HNSCC) by promoting antigen presentation and immune activation in the tumor microenvironment.

Patients and methods: The ALPHA study (NCT03695510) was a single-arm, Phase II study with Simon's 2-stage design. Afatinib and pembrolizumab were administered to patients with platinum-refractory, recurrent, or metastatic HNSCC. The primary endpoint was the objective response rate (ORR). The study applied gene expression analysis using a NanoString PanCancer Immune Profiling Panel and next-generation sequencing using FoundationOne CDx.

Results: From January 2019 to March 2020, the study enrolled 29 eligible patients. Common treatment-related adverse events were skin rash (75.9%), diarrhea (58.6%), and paronychia (44.8%). Twelve patients (41.4%) had an objective partial response to treatment. The median progression-free survival was 4.1 months, and the median overall survival was 8.9 months. In a paired tissue analysis, afatinib-pembrolizumab were found to upregulate genes involved in antigen presentation, immune activation, and natural killer cell-mediated cytotoxicity. Unaltered methylthioadenosine phosphorylase and EGFR amplification may predict the clinical response to the therapy.

Conclusions: Afatinib may augment pembrolizumab therapy and improve the ORR in patients with HNSCC. Bioinformatics analysis suggested the enhancement of antigen presentation machinery in the tumor microenvironment.

Trial registration: ClinicalTrials.gov NCT03695510 NCT04839471.

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

Figures

Figure 1.
Figure 1.
Clinical response to pembrolizumab–afatinib combined therapy in patients with HNSCC. A, Waterfall plot showing the best overall response (n = 29). B, Progression-free survival (n = 29). C, Overall survival (n = 29). D, The duration of response in responders (n = 12).
Figure 2.
Figure 2.
Paired tissue analysis. A, mRNA differential expression. Red dots represent genes with significant differences in mRNA expression (Benjamini–Yekutieli method, adjusted P < 0.05); B and C, Gene expression differences between paired pre- and post-treatment biopsy samples. *, P < 0.05; **, P < 0.01; ***, P < 0.001. D, Gene network analysis (by StringApp), showing the network of the upregulated genes in the tumor microenvironment. E, Gene set enrichment analysis of paired tissue mRNA samples. In the GO Biological Process analysis, only the top 10 downregulated gene sets are listed. F, Leading-edge analysis of three gene sets with immune cell regulatory functions.
Figure 3.
Figure 3.
Targeted gene mutation analysis. A, Genes were selected if three or more patients had mutations. TMB, tumor mutational burden in mutations/megabase. Survival analysis according to MTAP status. B, Progression-free survival. C, Overall survival. D, Comparing patients with altered MTAP versus unaltered MTAP by gene set enrichment analysis. In the present study and TCGA HNSCC analyses, tumors with altered MTAP had a more suppressed microenvironment. E, CIBERSORTx analysis: In this study, patients with MTAP loss or mutation had a low fraction of CD8+ T cells in the tumor microenvironment (Mann–Whitney U test, P = 0.0037, FDR q = 0.08).
Figure 4.
Figure 4.
Differences in mutation allele frequency (MAF) between pre-treatment and post-progression specimens. A, Best overall response: partial response. B, Best overall response: partial response. In the post-progression biopsy, a new MTAP loss and a new CDKN2A/B loss were detected. C, The patient had a good initial response to treatment. However, tumor regrowth occurred rapidly before the first imaging assessment. The best overall response of the patient was disease progression.

References

    1. Ferris RL, Blumenschein G Jr, Fayette J, Guigay J, Colevas AD, Licitra L, et al. . Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med 2016;375:1856–67.
    1. Cohen EEW, Soulières D, Le Tourneau C, Dinis J, Licitra L, Ahn MJ, et al. . Pembrolizumab versus methotrexate, docetaxel, or cetuximab for recurrent or metastatic head-and-neck squamous cell carcinoma (KEYNOTE-040): a randomized, open-label, phase 3 study. Lancet 2019;393:156–67.
    1. Burtness B, Harrington KJ, Greil R, Soulières D, Tahara M, de Castro G Jr, et al. . Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): a randomised, open-label, phase 3 study. Lancet 2019;394:1915–28.
    1. Kalbasi A, Ribas A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat Rev Immunol 2020;20:25–39.
    1. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, et al. . PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014;515:568–71.
    1. Jansen CS, Prokhnevska N, Master VA, Sanda MG, Carlisle JW, Bilen MA, et al. . An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature 2019;576:465–70.
    1. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity 2013;39:1–10.
    1. Basham TY, Merigan TC. Recombinant interferon-gamma increases HLA-DR synthesis and expression. J Immunol 1983;130:1492–4.
    1. Lee JH, Shklovskaya E, Lim SY, Carlino MS, Menzies AM, Stewart A, et al. . Transcriptional downregulation of MHC class I and melanoma de- differentiation in resistance to PD-1 inhibition. Nat Commun 2020;11:1897.
    1. Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, et al. . Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 2016;375:819–29.
    1. Concha-Benavente F, Srivastava RM, Trivedi S, Lei Y, Chandran U, Seethala RR, et al. . Identification of the cell-intrinsic and -extrinsic pathways downstream of EGFR and IFNγ that induce PD-L1 expression in head and neck cancer. Cancer Res 2016;76:1031–43.
    1. Leibowitz MS, Srivastava RM, Andrade Filho PA, Egloff AM, Wang L, Seethala RR, et al. . SHP2 is overexpressed and inhibits pSTAT1-mediated APM component expression, T-cell attracting chemokine secretion, and CTL recognition in head and neck cancer cells. Clin Cancer Res 2013;19:798–808.
    1. Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell 2015;28:690–714.
    1. Gandhi L, Rodríguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. . Pembrolizumab plus chemotherapy in metastatic non–small cell lung cancer. N Engl J Med 2018;378:2078–92.
    1. Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, et al. . First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial. Lancet 2021;398:27–40.
    1. Lizotte PH, Hong RL, Luster TA, Cavanaugh ME, Taus LJ, Wang S, et al. . A high-throughput immune-oncology screen identifies EGFR inhibitors as potent enhancers of antigen-specific cytotoxic t-lymphocyte tumor cell killing. Cancer Immunol Res 2018;6:1511–23.
    1. Machiels JP, Haddad RI, Fayette J, Licitra LF, Tahara M, Vermorken JB, et al. . Afatinib versus methotrexate as second-line treatment in patients with recurrent or metastatic squamous-cell carcinoma of the head and neck progressing on or after platinum-based therapy (LUX-Head & Neck 1): an open-label, randomised phase 3 trial. Lancet Oncol 2015;16:583–94.
    1. Guo Y, Ahn MJ, Chan A, Wang CH, Kang JH, Kim SB, et al. . Afatinib versus methotrexate as second-line treatment in Asian patients with recurrent or metastatic squamous cell carcinoma of the head and neck progressing on or after platinum-based therapy (LUX-Head & Neck 3): an open-label, randomised phase III trial. Ann Oncol 2019;30:1831–9.
    1. Geiss GK, Bumgarner RE, Birditt B, Dahl T, Dowidar N, Dunaway DL, et al. . Direct multiplexed measurement of gene expression with color-coded probe pairs. Nat Biotechnol 2008;26:317–25.
    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. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. . Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 2003;13:2498–504.
    1. Merico D, Isserlin R, Stueker O, Emili A, Bader GD. Enrichment map: a network-based method for gene-set enrichment visualization and interpretation. PLoS One 2010;5:e13984.
    1. Doncheva NT, Morris JH, Gorodkin J, Jensen LJ. Cytoscape StringApp: network analysis and visualization of proteomics data. J Proteome Res 2019;18:623–32.
    1. Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. . Robust enumeration of cell subsets from tissue expression profiles. Nat Methods 2015;12:453–7.
    1. Frampton GM, Fichtenholtz A, Otto GA, Wang K, Downing SR, He J, et al. . Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol 2013;31:1023–31.
    1. Hoadley KA, Yau C, Hinoue T, Wolf DM, Lazar AJ, Drill E, et al. . Cell-of-origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancer. Cell 2018;173:291–304.
    1. Emancipator K, Huang L, Aurora-Garg D, Bal T, Cohen EEW, Harrington K, et al. . Comparing programmed death ligand 1 scores for predicting pembrolizumab efficacy in head and neck cancer. Mod Pathol 2021;34:532–41.
    1. Litchfield K, Reading JL, Puttick C, Thakkar K, Abbosh C, Bentham R, et al. . Meta-analysis of tumor- and T-cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell 2021;184:596–614.
    1. House IG, Savas P, Lai J, Chen AXY, Oliver AJ, Teo ZL, et al. . Macrophage-derived CXCL9 and CXCL10 are required for antitumor immune responses following immune checkpoint blockade. Clin Cancer Res 2020;26:487–504.
    1. Barry KC, Hsu J, Broz ML, Cueto FJ, Binnewies M, Combes AJ, et al. . A natural killer-dendritic cell axis defines checkpoint therapy-responsive tumor microenvironments. Nat Med 2018;24:1178–91.
    1. Lee JJ, Kao KC, Chiu YL, Jung CJ, Liu CJ, Cheng SJ, et al. . Enrichment of human CCR6+ regulatory T cells with superior suppressive activity in oral cancer. J Immunol 2017;199:467–76.
    1. Barkal AA, Brewer RE, Markovic M, Kowarsky M, Barkal SA, Zaro BW, et al. . CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy. Nature 2019;572:392–6.
    1. Mowen KA, Tang J, Zhu W, Schurter BT, Shuai K, Herschman HR, et al. . Arginine methylation of STAT1 modulates IFNalpha/beta-induced transcription. Cell 2001;104:731–41.
    1. Gao J, Shi LZ, Zhao H, Chen J, Xiong L, He Q, et al. . Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti–CTLA-4 therapy. Cell 2016;167:397–404.
    1. Sacco AG, Chen R, Worden FP, Wong DJL, Adkins D, Swiecicki P, et al. . Pembrolizumab plus cetuximab in patients with recurrent or metastatic head and neck squamous cell carcinoma: an open-label, multi-arm, non-randomised, multicentre, phase 2 trial. Lancet Oncol 2021;22:883–92.
    1. Taylor MH, Lee CH, Makker V, Rasco D, Dutcus CE, Wu J, et al. . Phase IB/II trial of lenvatinib plus pembrolizumab in patients with advanced renal cell carcinoma, endometrial cancer, and other selected advanced solid tumors. J Clin Oncol 2020;38:1154–63.
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Lao CD, et al. . Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 2019;381:1535–46.
    1. Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, et al. . Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med 2019;380:1103–15.
    1. Hellmann MD, Paz-Ares L, Bernabe Caro R, Zurawski B, Kim SW, Carcereny Costa E, et al. . Nivolumab plus ipilimumab in advanced non–small cell lung cancer. N Engl J Med 2019;381:2020–31.
    1. Ferris RL, Haddad R, Even C, Tahara M, Dvorkin M, Ciuleanu TE, et al. . Durvalumab with or without tremelimumab in patients with recurrent or metastatic head and neck squamous cell carcinoma: EAGLE, a randomized, open-label phase III study. Ann Oncol 2020;31:942–50.
    1. Argiris A, Harrington K, Tahara M, Ferris RL, Gillison M, Fayette J, et al. . LBA36 - Nivolumab (N) + ipilimumab (I) vs. EXTREME as first-line (1L) treatment (tx) for recurrent/metastatic squamous cell carcinoma of the head and neck (R/M SCCHN): final results of CheckMate 651. Ann Oncol 2021;32:S1283–346.
    1. Harrington KJ, Kong A, Mach N, Chesney JA, Fernandez BC, Rischin D, et al. . Talimogene laherparepvec and pembrolizumab in recurrent or metastatic squamous cell carcinoma of the head and neck (MASTERKEY-232): a multicenter, phase 1b study. Clin Cancer Res 2020;26:5153–61.
    1. Massarelli E, William W, Johnson F, Kies M, Ferrarotto R, Guo M, et al. . Combining immune checkpoint blockade and tumor-specific vaccine for patients with incurable human papillomavirus 16-related cancer: a phase 2 clinical trial. JAMA Oncol 2019;5:67–73.
    1. Rodriguez CP, Wu QV, Voutsinas J, Fromm JR, Jiang X, Pillarisetty VG, et al. . A phase II trial of pembrolizumab and vorinostat in recurrent metastatic head and neck squamous cell carcinomas and salivary gland cancer. Clin Cancer Res 2020;26:837–45.
    1. Uppaluri R, Campbell KM, Egloff AM, Zolkind P, Skidmore ZL, Nussenbaum B, et al. . Neoadjuvant and adjuvant pembrolizumab in resectable locally advanced, human papillomavirus-unrelated head and neck cancer: a multicenter, phase II trial. Clin Cancer Res 2020;26:5140–52.
    1. Cristescu R, Mogg R, Ayers M, Albright A, Murphy E, Yearley J, et al. . Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 2018;362:eaar3593.
    1. Tu HF, Ko CJ, Lee CT, Lee CF, Lan SW, Lin HH, et al. . Afatinib exerts immunomodulatory effects by targeting the pyrimidine biosynthesis enzyme CAD. Cancer Res 2021;81:3270–82.
    1. Yang JC, Shepherd FA, Kim DW, Lee GW, Lee JS, Chang GC, et al. . Osimertinib plus durvalumab versus osimertinib monotherapy in EGFR T790M-positive NSCLC following previous EGFR TKI therapy: CAURAL brief report. J Thorac Oncol 2019;14:933–9.
    1. Yang JC, Gadgeel SM, Sequist LV, Wu CL, Papadimitrakopoulou VA, Su WC, et al. . Pembrolizumab in combination with erlotinib or gefitinib as first-line therapy for advanced NSCLC with sensitizing EGFR mutation. J Thorac Oncol 2019;14:553–9.
    1. Khoja L, Day D, Chen TWW, Siu LL, Hansen AR., Tumour- and class-specific patterns of immune-related adverse events of immune checkpoint inhibitors: a systematic review. Ann Oncol 2017;28:2377–85.
    1. Gohil SH, Iorgulescu JB, Braun DA, Keskin DB, Livak KJ. Applying high-dimensional single-cell technologies to the analysis of cancer immunotherapy. Nat Rev Clin Oncol 2021;18:244–56.
    1. Lundberg E, Borner GH. Spatial proteomics: a powerful discovery tool for cell biology. Nat Rev Mol Cell Biol 2019;20:285–302.
    1. Armingol E, Officer A, Harismendy O, Lewis NE., Deciphering cell–cell interactions and communication from gene expression. Nat Rev Genet 2021;22:71–88.

Source: PubMed

3
Tilaa