Association of Baseline and Pharmacodynamic Biomarkers With Outcomes in Patients Treated With the PD-1 Inhibitor Budigalimab

Stacie L Lambert, Chun Zhang, Claire Guo, Tolga Turan, David L Masica, Stefan Englert, Yuni Fang, James Sheridan, Robert Tyler McLaughlin, Catherine Tribouley, Greg Vosganian, Daniel Afar, Stacie L Lambert, Chun Zhang, Claire Guo, Tolga Turan, David L Masica, Stefan Englert, Yuni Fang, James Sheridan, Robert Tyler McLaughlin, Catherine Tribouley, Greg Vosganian, Daniel Afar

Abstract

Budigalimab, a novel anti-PD-1 monoclonal antibody, demonstrated efficacy and biomarker pharmacodynamics in patients with head and neck squamous cell carcinoma (HNSCC) or non-small cell lung cancer (NSCLC) consistent with those reported by other PD-1 inhibitors. Herein are presented additional outcomes of biomarker analyses from the phase 1 study of budigalimab monotherapy in patients with HNSCC and NSCLC (NCT03000257). PD-1 inhibitor naive patients with advanced HNSCC (n=41) or NSCLC (n=40) received budigalimab intravenously at 250 mg every 2 weeks (Q2W) or 500 mg Q4W until progression. Archival tumor specimens were evaluated by immunohistochemistry for CD8 and tumor PD-1 ligand 1 (PD-L1) expression, RNA, and whole-exome sequencing. Serum and whole blood samples were acquired at baseline and at select on-treatment time points. As of October 2019, best overall response of 15% in HNSCC and 18% in NSCLC was observed in all treated patients; both cohorts reported responses in PD-L1+ and PD-L1- tumors. Treatment with budigalimab was associated with increases in multiple soluble biomarkers including interferon gamma-induced chemokines. Expanded overall T-cell counts, total CD8 T-cell counts, and percentages of CD8+CD45RA-CD62L- effector memory T cells were observed at cycle 1, day 15 in responders. Univariate analysis demonstrated an association between prolonged progression-free survival and higher tumor mutational burden/neoantigen load, smaller tumor size, lower platelet-lymphocyte ratios, lower CCL23, lower colony-stimulating factor 1, and lower interleukin-6 levels at baseline. The biomarker analysis presented herein identified additional early pharmacodynamic biomarkers associated with anti-PD-1 activity and improved clinical responses to budigalimab in patients with advanced HNSCC and NSCLC.

Conflict of interest statement

AbbVie Inc. provided financial support for the study (NCT03000257) and participated in the design, study conduct, as well as the writing, review, and approval of the manuscript. Data analysis and interpretation was conducted by employees of AbbVie Inc.

S.L.L., C.Z., C.G., T.T., D.L.M., S.E., Y.F., J.S., R.T.M., C.T., and D.A.: AbbVie employees and may own stock. G.V.: former employee of AbbVie and may own stock.

Copyright © 2022 The Author(s). Published by Wolters Kluwer Health, Inc.

Figures

FIGURE 1
FIGURE 1
Immunohistochemical detection of PD-L1 percentage positive in tumor cells (A), PD-L1 intensity in inflammatory cells (B), and CD8 percentage positive (C) expression in archival tumor samples of patients with HNSCC and NSCLC treated with budigalimab monotherapy. A total of 71 patients (38 HNSCC and 33 NSCLC) were included in these analyses. Purple: EGFR mutated; blue: ALK rearrangement. + indicates positive; CR, complete response; HNSCC, head and neck squamous cell carcinoma; iPR, immune partial response; NSCLC, non–small cell lung cancer; PD, progressive disease; PD-L1, programmed cell death protein 1 ligand 1; PR, partial response; SD, stable disease.
FIGURE 2
FIGURE 2
Exome sequencing showed a relationship between tumor mutational burden (TMB) and best overall response (A); weak association of TMB with PFS was also observed, with a hazard ratio of 0.77 and a P-value of 0.126 (B). The optimized (sensitivity plus specificity) cutoff of 4.895, shown by the solid line, and an optimized (maximal sensitivity) cutoff of 2.47, shown by the dotted line were determined by performance as shown in Supplemental Digital Content Figure 1 (Supplemental Digital Content 1, http://links.lww.com/JIT/A666). A total of 32 patients (22 HNSCC and 10 NSCLC) were included in these analyses. BOR indicates best overall response; CR, complete response; HNSCC, head and neck squamous cell carcinoma; iPR, immune partial response; mono, monotherapy; NSCLC, non–small cell lung cancer; PD, progressive disease; PFS, progression-free survival; PR, partial response; SD, stable disease.
FIGURE 3
FIGURE 3
RNAseq analysis of the ∼40,000 transcripts that are nonzero in at least one sample detected multiple unique features with significant expression differences between responders and nonresponders in the indication-specific HNSCC, n=32 (A) and NSCLC, n=12 (B) data sets, as well as the combined (C) data set, n=44. Orange and green dots represent significantly differentially expressed transcripts (|FC| >2, P-value <0.05, FDR <0.25), with orange indicating higher expression in responders and green indicating lower expression in responders. Qiagen Ingenuity Pathway Analysis of differential gene expression by responders and nonresponders showed significantly upregulated pathways (orange) and downregulated pathways (blue) in responding patients with HNSCC (D). In the combined data set, EDIL3 was associated with both BOR (E) and PFS (F). AUC indicates area under the concentration-time curve; BOR, best overall response; CR, complete response; FC, fold change; FDR, false discovery rate; HNSCC, head and neck squamous cell carcinoma; HR, hazard ratio; iPR, immune partial response; NSCLC, non–small cell lung cancer; PD, progressive disease; PFS, progression-free survival; PR, partial response; RNAseq, RNA sequencing; SD, stable disease.
FIGURE 4
FIGURE 4
Immunophenotyping detected pharmacodynamic changes induced by budigalimab, including the percentages of Ki67+ CD4 and CD8 T cells [mean and SE shown from a total of 44 patients (24 HNSCC and 20 NSCLC) with baseline and postbaseline Ki67 assessments, with 9 responders] (A), circulating cell counts of CD3–, CD4–, and CD8+ T-cell counts (mean and SE shown from a total of 71 patients with baseline and postbaseline total T cells, helper T cells, cytotoxic T cells, B cells, and NK cells assessments, with 13 responders) (B), and percentages of CD8 T-cell subsets on the basis of CD28 and CD95 expression (mean and SE shown from a total of 49 patients with baseline and postbaseline memory subset assessments, with 10 responders) (C). – indicates negative; +, positive; C, cycle; D, day; HNSCC, head and neck squamous cell carcinoma; mem, memory; NK, natural killer; NSCLC, non–small cell lung cancer.
FIGURE 5
FIGURE 5
Budigalimab treatment induced soluble biomarkers of T-cell activity associated with differential response. Data shown are from a total of 65 patients with baseline and postbaseline assessments including 12 responders and 43 nonresponders. Longitudinal analysis of significantly upregulated biomarkers across all patients shows IFN-γ peak expression at the C1D2 time point (A), while CXCL9 (MIG), CXCL10, CXCL11, GZMH, TNFRSF4, and TNFRSF9 peak at the C2D1 time point (B, C). Significant PFS differences were observed between patients with the highest and lowest induction at C1D2 of IFN-γ (D), IL-8 (E), or CXCL9 (MIG) (F). C indicates cycle; D, day; FC, fold change; HSNCC, head and neck squamous cell carcinoma; IFN, interferon; IL, interleukin; INF, inflammatory; IO, immuno-oncologic; MIG, mitogen-inducible gene; NSCLC, non–small cell lung cancer; TNF, tumor necrosis factor.

References

    1. Weng YM, Peng M, Hu MX, et al. . Clinical and molecular characteristics associated with the efficacy of PD-1/PD-L1 inhibitors for solid tumors: a meta-analysis. Onco Targets Ther. 2018;11:7529–7542.
    1. Prasad V, Kaestner V. Nivolumab and pembrolizumab: monoclonal antibodies against programmed cell death-1 (PD-1) that are interchangeable. Semin Oncol. 2017;44:132–135.
    1. Ancevski Hunter K, Socinski MA, Villaruz LC. PD-L1 testing in guiding patient selection for PD-1/PD-L1 inhibitor therapy in lung cancer. Mol Diagn Ther. 2018;22:1–10.
    1. Voong KR, Feliciano J, Becker D, et al. . Beyond PD-L1 testing—emerging biomarkers for immunotherapy in non-small cell lung cancer. Ann Transl Med. 2017;5:376.
    1. Ayers M, Lunceford J, Nebozhyn M, et al. . IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest. 2017;127:2930–2940.
    1. Prat A, Navarro A, Paré L, et al. . Immune-related gene expression profiling after PD-1 blockade in non-small cell lung carcinoma, head and neck squamous cell carcinoma, and melanoma. Cancer Res. 2017;77:3540–3550.
    1. Yarchoan M, Albacker LA, Hopkins AC, et al. . PD-L1 expression and tumor mutational burden are independent biomarkers in most cancers. JCI Insight. 2019;4:e126908.
    1. Zheng H, Liu X, Zhang J, et al. . Expression of PD-1 on CD4+ T cells in peripheral blood associates with poor clinical outcome in non-small cell lung cancer. Oncotarget. 2016;7:56233–56240.
    1. Italiano A, Cassier PA, Lin CC, et al. . First-in-human phase 1 study of budigalimab, an anti-PD-1 inhibitor, in patients with non-small cell lung cancer and head and neck squamous cell carcinoma. Cancer Immunol Immunother. 2021. Available at: .
    1. Seymour L, Bogaerts J, Perrone A, et al. . iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017;18:e143–e152.
    1. Garon EB, Rizvi NA, Hui R, et al. . Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372:2018–2028.
    1. Gettinger SN, Horn L, Gandhi L, et al. . Overall survival and long-term safety of nivolumab (anti-programmed death 1 antibody, BMS-936558, ONO-4538) in patients with previously treated advanced non-small-cell lung cancer. J Clin Oncol. 2015;33:2004–2012.
    1. Seiwert TY, Burtness B, Mehra R, et al. . Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): an open-label, multicentre, phase 1b trial. Lancet Oncol. 2016;17:956–965.
    1. Vaidyanathan R, Soon RH, Zhang P, et al. . Cancer diagnosis: from tumor to liquid biopsy and beyond. Lab Chip. 2018;19:11–34.
    1. Sanmamed MF, Perez-Gracia JL, Schalper KA, et al. . Changes in serum interleukin-8 (IL-8) levels reflect and predict response to anti-PD-1 treatment in melanoma and non-small-cell lung cancer patients. Ann Oncol. 2017;28:1988–1995.
    1. Juliá EP, Mandó P, Rizzo MM, et al. . Peripheral changes in immune cell populations and soluble mediators after anti-PD-1 therapy in non-small cell lung cancer and renal cell carcinoma patients. Cancer Immunol Immunother. 2019;68:1585–1596.
    1. Hendrickx W, Simeone I, Anjum S, et al. . Identification of genetic determinants of breast cancer immune phenotypes by integrative genome-scale analysis. Oncoimmunology. 2017;6:e1253654.
    1. Brahmer JR, Drake CG, Wollner I, et al. . Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28:3167–3175.
    1. Das R, Verma R, Sznol M, et al. . Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J Immunol. 2015;194:950–959.
    1. Choueiri TK, Fishman MN, Escudier B, et al. . Immunomodulatory activity of nivolumab in metastatic renal cell carcinoma. Clin Cancer Res. 2016;22:5461–5471.
    1. Herbst RS, Soria JC, Kowanetz M, et al. . Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–567.
    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;348:124–128.
    1. Cristescu R, Mogg R, Ayers M, et al. . Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science. 2018;362:eaar3593.
    1. Hu-Lieskovan S, Lisberg A, Zaretsky JM, et al. . Tumor characteristics associated with benefit from pembrolizumab in advanced non-small cell lung cancer. Clin Cancer Res. 2019;25:5061–5068.
    1. Berghoff AS, Bellosillo B, Caux C, et al. . Immune checkpoint inhibitor treatment in patients with oncogene-addicted non-small cell lung cancer (NSCLC): summary of a multidisciplinary round-table discussion. ESMO Open. 2019;4:e000498.
    1. Gameiro SF, Ghasemi F, Barrett JW, et al. . Treatment-naïve HPV+ head and neck cancers display a T-cell-inflamed phenotype distinct from their HPV- counterparts that has implications for immunotherapy. Oncoimmunology. 2018;7:e1498439.
    1. Harjunpää H, Asens ML, Guenther C, et al. . Cell adhesion molecules and their roles and regulation in the immune and tumor microenvironment. Front Immunol. 2019;10:1078.
    1. Jeong D, Ban S, Oh S, et al. . Prognostic significance of EDIL3 expression and correlation with mesenchymal phenotype and microvessel density in lung adenocarcinoma. Sci Rep. 2017;7:8649.
    1. Argyle D, Kitamura T. Targeting macrophage-recruiting chemokines as a novel therapeutic strategy to prevent the progression of solid tumors. Front Immunol. 2018;9:2629.
    1. Diem S, Schmid S, Krapf M, et al. . Neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) as prognostic markers in patients with non-small cell lung cancer (NSCLC) treated with nivolumab. Lung Cancer. 2017;111:176–181.
    1. Song X, Chen D, Yuan M, et al. . Total lymphocyte count, neutrophil-lymphocyte ratio, and platelet-lymphocyte ratio as prognostic factors in advanced non-small cell lung cancer with chemoradiotherapy. Cancer Manag Res. 2018;10:6677–6683.
    1. Choudhary MM, France TJ, Teknos TN, et al. . Interleukin-6 role in head and neck squamous cell carcinoma progression. World J Otorhinolaryngol Head Neck Surg. 2016;2:90–97.
    1. Silva EM, Mariano VS, Pastrez PRA, et al. . High systemic IL-6 is associated with worse prognosis in patients with non-small cell lung cancer. PLoS One. 2017;12:e0181125.
    1. Zhu Y, Yang J, Xu D, et al. . Disruption of tumour-associated macrophage trafficking by the osteopontin-induced colony-stimulating factor-1 signalling sensitizes hepatocellular carcinoma to anti-PD-L1 blockade. Gut. 2019;68:1653–1666.
    1. Yost KE, Satpathy AT, Wells DK, et al. . Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med. 2019;25:1251–1259.
    1. Valpione S, Galvani E, Tweedy J, et al. . Immune-awakening revealed by peripheral T cell dynamics after one cycle of immunotherapy. Nat Cancer. 2020;1:210–221.
    1. Huang AC, Postow MA, Orlowski RJ, et al. . T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature. 2017;545:60–65.
    1. Kamphorst AO, Pillai RN, Yang S, et al. . Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci U S A. 2017;114:4993–4998.
    1. Cheng HW, Onder L, Cupovic J, et al. . CCL19-producing fibroblastic stromal cells restrain lung carcinoma growth by promoting local antitumor T-cell responses. J Allergy Clin Immunol. 2018;142:1257–1271.
    1. Bremnes RM, Camps C, Sirera R. Angiogenesis in non-small cell lung cancer: the prognostic impact of neoangiogenesis and the cytokines VEGF and bFGF in tumours and blood. Lung Cancer. 2006;51:143–158.
    1. Khademi B, Soleimanpour M, Ghaderi A, et al. . Prognostic and predictive value of serum vascular endothelial growth factor (VEGF) in squamous cell carcinoma of the head and neck. Oral Maxillofac Surg. 2014;18:187–196.
    1. Mehta A, Rucevic M, Sprecher E, et al. . The use of plasma proteomic markers to understand the biology of immunotherapy response. J Clin Oncol. 2020;38(15 suppl):517s.
    1. Chow MT, Ozga AJ, Servis RL, et al. . Intratumoral activity of the CXCR3 chemokine system is required for the efficacy of anti-PD-1 therapy. Immunity. 2019;50:1498–1512.
    1. Oyanagi J, Koh Y, Sato K, et al. . Predictive value of serum protein levels in patients with advanced non-small cell lung cancer treated with nivolumab. Lung Cancer. 2019;132:107–113.
    1. Subrahmanyam PB, Dong Z, Gusenleitner D, et al. . Distinct predictive biomarker candidates for response to anti-CTLA-4 and anti-PD-1 immunotherapy in melanoma patients. J Immunother Cancer. 2018;6:18.
    1. Kim KH, Cho J, Ku BM, et al. . The first-week proliferative response of peripheral blood PD-1+CD8+ T cells predicts the response to anti-PD-1 therapy in solid tumors. Clin Cancer Res. 2019;25:2144–2154.

Source: PubMed

Подписаться