The Combined Effect of FGFR Inhibition and PD-1 Blockade Promotes Tumor-Intrinsic Induction of Antitumor Immunity

Treatments that use a pan-FGFR (fibroblast growth factor receptor) inhibitor plus anti–PD-1 can boost antitumor responses in tumors harboring genetic mutations in driver oncogenes. The agents remodel the tumor microenvironment and enhance the expansion of T-cell clones. The success of targeted or immune therapies is often hampered by the emergence of resistance and/or clinical benefit in only a subset of patients. We hypothesized that combining targeted therapy with immune modulation would show enhanced antitumor responses. Here, we explored the combination potential of erdafitinib, a fibroblast growth factor receptor (FGFR) inhibitor under clinical development, with PD-1 blockade in an autochthonous FGFR2K660N/p53mut lung cancer mouse model. Erdafitinib monotherapy treatment resulted in substantial tumor control but no significant survival benefit. Although anti–PD-1 alone was ineffective, the erdafitinib and anti–PD-1 combination induced significant tumor regression and improved survival. For both erdafitinib monotherapy and combination treatments, tumor control was accompanied by tumor-intrinsic, FGFR pathway inhibition, increased T-cell infiltration, decreased regulatory T cells, and downregulation of PD-L1 expression on tumor cells. These effects were not observed in a KRASG12C-mutant genetically engineered mouse model, which is insensitive to FGFR inhibition, indicating that the immune changes mediated by erdafitinib may be initiated as a consequence of tumor cell killing. A decreased fraction of tumor-associated macrophages also occurred but only in combination-treated tumors. Treatment with erdafitinib decreased T-cell receptor (TCR) clonality, reflecting a broadening of the TCR repertoire induced by tumor cell death, whereas combination with anti–PD-1 led to increased TCR clonality, suggesting a more focused antitumor T-cell response. Our results showed that the combination of erdafitinib and anti–PD-1 drives expansion of T-cell clones and immunologic changes in the tumor microenvironment to support enhanced antitumor immunity and survival.

[1]  Xiaoting Yu,et al.  Assessing Therapeutic Efficacy of MEK Inhibition in a KRASG12C-Driven Mouse Model of Lung Cancer , 2018, Clinical Cancer Research.

[2]  Stephen Broderick,et al.  Neoadjuvant PD‐1 Blockade in Resectable Lung Cancer , 2018, The New England journal of medicine.

[3]  Tim Beißbarth,et al.  From somatic variants towards precision oncology: Evidence-driven reporting of treatment options in molecular tumor boards , 2018, Genome Medicine.

[4]  T. Gajewski,et al.  Impact of oncogenic pathways on evasion of antitumour immune responses , 2018, Nature Reviews Cancer.

[5]  R. Emerson,et al.  Radiotherapy and CTLA-4 Blockade Shape the TCR Repertoire of Tumor-Infiltrating T Cells , 2017, Cancer Immunology Research.

[6]  T. Bivona,et al.  Understanding and targeting resistance mechanisms in NSCLC , 2017, Nature Reviews Cancer.

[7]  William B. Goggins,et al.  Incidence and mortality of lung cancer: global trends and association with socioeconomic status , 2017, Scientific Reports.

[8]  N. Turner,et al.  Advances and challenges in targeting FGFR signalling in cancer , 2017, Nature Reviews Cancer.

[9]  A. Santiago-Walker,et al.  Oncogenic Characterization and Pharmacologic Sensitivity of Activating Fibroblast Growth Factor Receptor (FGFR) Genetic Alterations to the Selective FGFR Inhibitor Erdafitinib , 2017, Molecular Cancer Therapeutics.

[10]  C. Murray,et al.  Discovery and Pharmacological Characterization of JNJ-42756493 (Erdafitinib), a Functionally Selective Small-Molecule FGFR Family Inhibitor , 2017, Molecular Cancer Therapeutics.

[11]  W. Cho,et al.  Precision medicine in immune checkpoint blockade therapy for non-small cell lung cancer , 2017, Clinical and Translational Medicine.

[12]  E. Mardis,et al.  Temporally Distinct PD-L1 Expression by Tumor and Host Cells Contributes to Immune Escape , 2017, Cancer Immunology Research.

[13]  F. Safa,et al.  Anti–PD-1/PD-L1 antibodies in non-small cell lung cancer: the era of immunotherapy , 2017, Expert review of anticancer therapy.

[14]  H. Ishwaran,et al.  Tumor Interferon Signaling Regulates a Multigenic Resistance Program to Immune Checkpoint Blockade , 2016, Cell.

[15]  M. Stanford,et al.  Anti-PD-1 increases the clonality and activity of tumor infiltrating antigen specific T cells induced by a potent immune therapy consisting of vaccine and metronomic cyclophosphamide , 2016, Journal of Immunotherapy for Cancer.

[16]  Adam P. Rosebrock,et al.  Mutational landscape of EGFR-, MYC-, and Kras-driven genetically engineered mouse models of lung adenocarcinoma , 2016, Proceedings of the National Academy of Sciences.

[17]  J. Blay,et al.  Safety and activity of the pan–fibroblast growth factor receptor (FGFR) inhibitor erdafitinib in phase 1 study patients with advanced urothelial carcinoma , 2016 .

[18]  Ryan L. Kelly,et al.  Eradication of large established tumors in mice by combination immunotherapy that engages innate and adaptive immune responses , 2016, Nature Medicine.

[19]  S. Gettinger,et al.  PD-1 Axis Inhibitors in EGFR- and ALK-Driven Lung Cancer: Lost Cause? , 2016, Clinical Cancer Research.

[20]  Lawren C. Wu,et al.  Targeted Therapy and Checkpoint Immunotherapy Combinations for the Treatment of Cancer. , 2016, Trends in immunology.

[21]  M. Katoh FGFR inhibitors: Effects on cancer cells, tumor microenvironment and whole-body homeostasis (Review) , 2016, International journal of molecular medicine.

[22]  S. Digumarthy,et al.  EGFR Mutations and ALK Rearrangements Are Associated with Low Response Rates to PD-1 Pathway Blockade in Non–Small Cell Lung Cancer: A Retrospective Analysis , 2016, Clinical Cancer Research.

[23]  G. Steinberg,et al.  Molecular Drivers of the Non–T-cell-Inflamed Tumor Microenvironment in Urothelial Bladder Cancer , 2016, Cancer Immunology Research.

[24]  Jing Wang,et al.  STK11/LKB1 Deficiency Promotes Neutrophil Recruitment and Proinflammatory Cytokine Production to Suppress T-cell Activity in the Lung Tumor Microenvironment. , 2016, Cancer research.

[25]  C. Jiang,et al.  Regulation of PD-L1: a novel role of pro-survival signalling in cancer. , 2016, Annals of oncology : official journal of the European Society for Medical Oncology.

[26]  R. Wilkinson,et al.  Transformation of the tumour microenvironment by a CD40 agonist antibody correlates with improved responses to PD-L1 blockade in a mouse orthotopic pancreatic tumour model , 2016, Oncotarget.

[27]  R. Weissleder,et al.  Immunogenic Chemotherapy Sensitizes Tumors to Checkpoint Blockade Therapy. , 2016, Immunity.

[28]  M. Vignali,et al.  T‐cell receptor profiling in cancer , 2015, Molecular oncology.

[29]  J. Tabernero,et al.  Phase I Dose-Escalation Study of JNJ-42756493, an Oral Pan-Fibroblast Growth Factor Receptor Inhibitor, in Patients With Advanced Solid Tumors. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[30]  T. Gajewski,et al.  Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity , 2015, Nature.

[31]  Martin L. Miller,et al.  Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer , 2015, Science.

[32]  William S. DeWitt,et al.  Dynamics of the Cytotoxic T Cell Response to a Model of Acute Viral Infection , 2015, Journal of Virology.

[33]  R. Emerson,et al.  PD-1 blockade induces responses by inhibiting adaptive immune resistance , 2014, Nature.

[34]  M. Meyerson,et al.  Kinase domain activation of FGFR2 yields high-grade lung adenocarcinoma sensitive to a Pan-FGFR inhibitor in a mouse model of NSCLC. , 2014, Cancer research.

[35]  Steven J. M. Jones,et al.  Comprehensive molecular profiling of lung adenocarcinoma , 2014, Nature.

[36]  Yuquan Wei,et al.  Reductions in Myeloid-Derived Suppressor Cells and Lung Metastases using AZD4547 Treatment of a Metastatic Murine Breast Tumor Model , 2014, Cellular Physiology and Biochemistry.

[37]  Yuquan Wei,et al.  Inhibition of FGFR signaling by PD173074 improves antitumor immunity and impairs breast cancer metastasis , 2014, Breast Cancer Research and Treatment.

[38]  R. Emerson,et al.  Using synthetic templates to design an unbiased multiplex PCR assay , 2013, Nature Communications.

[39]  Jeanne Shen,et al.  Clinical, Pathologic, and Biologic Features Associated with BRAF Mutations in Non–Small Cell Lung Cancer , 2013, Clinical Cancer Research.

[40]  Laurence Zitvogel,et al.  Immunogenic cell death in cancer therapy. , 2013, Annual review of immunology.

[41]  Bruce E Johnson,et al.  The Introduction of Systematic Genomic Testing for Patients with Non–Small-Cell Lung Cancer , 2012, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[42]  T. Griffin,et al.  Fibroblast Growth Factor Receptor 1 Activation in Mammary Tumor Cells Promotes Macrophage Recruitment in a CX3CL1-Dependent Manner , 2012, PloS one.

[43]  C. Desmarais,et al.  Ultra-sensitive detection of rare T cell clones. , 2012, Journal of immunological methods.

[44]  J. Wesche,et al.  Fibroblast growth factors and their receptors in cancer. , 2011, The Biochemical journal.

[45]  I. Petersen,et al.  Frequent and Focal FGFR1 Amplification Associates with Therapeutically Tractable FGFR1 Dependency in Squamous Cell Lung Cancer , 2010, Science Translational Medicine.

[46]  A. Ashworth,et al.  FGFR1 amplification drives endocrine therapy resistance and is a therapeutic target in breast cancer. , 2010, Cancer research.

[47]  N. Turner,et al.  Fibroblast growth factor signalling: from development to cancer , 2010, Nature Reviews Cancer.

[48]  Hugo M. Horlings,et al.  Integrative molecular profiling of triple negative breast cancers identifies amplicon drivers and potential therapeutic targets , 2009, Oncogene.

[49]  Abigail Wacher,et al.  Comprehensive assessment of T-cell receptor beta-chain diversity in alphabeta T cells. , 2009, Blood.

[50]  T. Jacks,et al.  Conditional mouse lung cancer models using adenoviral or lentiviral delivery of Cre recombinase , 2009, Nature Protocols.

[51]  K. Sakaguchi,et al.  FRAG1, a gene that potently activates fibroblast growth factor receptor by C-terminal fusion through chromosomal rearrangement. , 1996, Proceedings of the National Academy of Sciences of the United States of America.