Antitumor immune effects of preoperative sitravatinib and nivolumab in oral cavity cancer: SNOW window-of-opportunity study

Marc Oliva, Douglas Chepeha, Daniel V Araujo, J Javier Diaz-Mejia, Peter Olson, Amy Prawira, Anna Spreafico, Scott V Bratman, Tina Shek, John de Almeida, Aaron R Hansen, Andrew Hope, David Goldstein, Ilan Weinreb, Stephen Smith, Bayardo Perez-Ordoñez, Jonathan Irish, Dax Torti, Jeffrey P Bruce, Ben X Wang, Anthony Fortuna, Trevor J Pugh, Hirak Der-Torossian, Ronald Shazer, Nickolas Attanasio, Qingyan Au, Antony Tin, Jordan Feeney, Himanshu Sethi, Alexey Aleshin, Isan Chen, Lillian Siu, Marc Oliva, Douglas Chepeha, Daniel V Araujo, J Javier Diaz-Mejia, Peter Olson, Amy Prawira, Anna Spreafico, Scott V Bratman, Tina Shek, John de Almeida, Aaron R Hansen, Andrew Hope, David Goldstein, Ilan Weinreb, Stephen Smith, Bayardo Perez-Ordoñez, Jonathan Irish, Dax Torti, Jeffrey P Bruce, Ben X Wang, Anthony Fortuna, Trevor J Pugh, Hirak Der-Torossian, Ronald Shazer, Nickolas Attanasio, Qingyan Au, Antony Tin, Jordan Feeney, Himanshu Sethi, Alexey Aleshin, Isan Chen, Lillian Siu

Abstract

Background: Sitravatinib, a tyrosine kinase inhibitor that targets TYRO3, AXL, MERTK and the VEGF receptor family, is predicted to increase the M1 to M2-polarized tumor-associated macrophages ratio in the tumor microenvironment and have synergistic antitumor activity in combination with anti-programmed death-1/ligand-1 agents. SNOW is a window-of-opportunity study designed to evaluate the immune and molecular effects of preoperative sitravatinib and nivolumab in patients with oral cavity squamous cell carcinoma.

Methods: Patients with newly-diagnosed untreated T2-4a, N0-2 or T1 >1 cm-N2 oral cavity carcinomas were eligible. All patients received sitravatinib 120 mg daily from day 1 up to 48 hours pre-surgery and one dose of nivolumab 240 mg on day 15. Surgery was planned between day 23 and 30. Standard of care adjuvant radiotherapy was given based on clinical stage. Tumor photographs, fresh tumor biopsies and blood samples were collected at baseline, at day 15 after sitravatinib alone, and at surgery after sitravatinib-nivolumab combination. Tumor flow cytometry, multiplex immunofluorescence staining and single-cell RNA sequencing (scRNAseq) were performed on tumor biopsies to study changes in immune-cell populations. Tumor whole-exome sequencing and circulating tumor DNA and cell-free DNA were evaluated at each time point.

Results: Ten patients were included. Grade 3 toxicity occurred in one patient (hypertension); one patient required sitravatinib dose reduction, and one patient required discontinuation and surgery delay due to G2 thrombocytopenia. Nine patients had clinical-to-pathological downstaging, with one complete response. Independent pathological treatment response (PTR) assessment confirmed a complete PTR and two major PTRs. With a median follow-up of 21 months, all patients are alive with no recurrence. Circulating tumor DNA and cell-free DNA dynamics correlated with clinical and pathological response and distinguished two patient groups with different tumor biological behavior after sitravatinib alone (1A) versus sitravatinib-nivolumab (1B). Tumor immunophenotyping and scRNAseq analyses revealed differential changes in the expression of immune cell populations and sitravatinib-targeted and hypoxia-related genes in group 1A vs 1B patients.

Conclusions: The SNOW study shows sitravatinib plus nivolumab is safe and leads to deep clinical and pathological responses in oral cavity carcinomas. Multi-omic biomarker analyses dissect the differential molecular effects of sitravatinib versus the sitravatinib-nivolumab and revealed patients with distinct tumor biology behavior.

Trial registration number: NCT03575598.

Keywords: clinical trials as topic; head and neck neoplasms; immunotherapy; macrophages; tumor biomarkers.

Conflict of interest statement

Competing interests: DVA: honoraria from GSK, MSD and Pfizer. AP: research funding from BMS. AS: consultant for (Advisory Board), Merck (compensated), Bristol-Myers Squibb (compensated), Novartis (compensated), Oncorus (compensated), Janssen (compensated). Grant/Research support from (Clinical Trials): Novartis, Bristol Myers Squibb, Symphogen, AstraZeneca/Medimmune, Merck, Bayer, Surface Oncology, Northern Biologics, Janssen Oncology/Johnson & Johnson, Roche, Regeneron, Alkermes, Array Biopharma/Pfizer, GSK. ARH: consultant for (Advisory Board) Bristol Myers Squibb (compensated), Merck (compensated), GlaxoSmithKline (compensated), Eisai (compensated). Grant/Research support from (Clinical Trials): Karyopharm, Bristol Myers Squibb, GlaxoSmithKline, Merck, Boerhinger-Ingelheim, AstraZeneca/Medimmune, Roche/Genentech, Macrogenics, Astellas Pharma, Janssen. PO, RS, HD are employees and shareholders of Mirati Therapeutics. NA, QA are employees of Neogenomics Laboratories. LS: consulting/advisory arrangements with Merck, Pfizer, Celgene, AstraZeneca, Morphosys, Roche, Oncorus, Symphogen, Seattle Genetics, GlaxoSmithKline, Voronoi, Arvinas, Tessa, Navire, Relay, Rubius, Janpix, Daiichi Sanyko; stock ownership of Agios (spouse); leadership position in Treadwell Therapeutics (spouse); and institution receives clinical trials support from Novartis, Bristol Myers Squibb, Pfizer, Boerhinger-Ingelheim, GlaxoSmithKline, Roche/Genentech, Karyopharm, AstraZeneca, Merck, Celgene, Astellas, Bayer, AbbVie, Amgen, Symphogen, Intensity Therapeutics, Mirati Therapeutics, Shattucks, Avid.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY. Published by BMJ.

Figures

Figure 1
Figure 1
ctDNA (MTM/mL) (A) and cfDNA (ng/mL) (B) dynamics following study treatment. (A) Two-dimensional line charts showing MTM/mL at each of the three time points. (B) Two-dimensional line charts showing cfDNA in ng/mL at each of the three time points. The symbols represent each individual patient. The colors represent the groups according to the distinct tumor biological behavior following treatment. cfDNA, cell-free DNA; ctDNA, circulating tumor DNA; D15, day 15; MTM, mean tumor molecules; PRE, pretreatment; SRG, pre-surgery.
Figure 2
Figure 2
ctDNA dynamics correlated with tumor changes following sitravatinib and sitravatinib plus nivolumab. Charts showing log-scale changes in ctDNA and cfDNA (Y-axis) at each time point (X-axis) in each individual patient. Tumor photographs performed during study at each of the corresponding time points are shown above the line charts for each patient. Arrows indicate the location of the primary tumor. ctDNA, circulating tumor DNA; cfDNA, cell-free DNA; D15, day 15; MTM, mean tumor molecules; PRE, pretreatment; SRG, pre-surgery.
Figure 3
Figure 3
Changes in tumor-associated macrophage populations following sitravatinib alone (D15) and sitravatinib plus nivolumab (SRG). (A) Uniform Manifold Approximation and Projection (UMAP) plots showing the integration of 15 samples from five SNOW patients at three time points. Colors represent cell types. (B) Multiplexing immuno-fluorescence staining in tumor biopsies at pre-treatment (PRE), day 15 (D15) and pre-surgery (SRG) using NeoGenomics MultiOmyx panels showing changes in macrophages subpopulations: M1 type (CD68+CD163–) shown in red, M2 type (CD68+CD163+) shown in yellow and M1 intermediate type (CD68+HLA-DR+CD163–) shown in magenta. Upper images show H&E staining of tissue sample. (C) Ratio of M1/M2 macrophages detected using IHC and scRNAseq measurements. Orange: 1A=responders to sitravatinib; blue: 1B=responders to sitravatinib–nivolumab; black=unclassifiable. IHC, immunohistochemistry; NK, natural killer; MDSC, myeloid-derived suppressor cell; scRNAseq, single-cell RNA sequencing.
Figure 4
Figure 4
Single-cell RNA sequencing pathways enrichment analysis comparing Group 1A versus Group 1B patients at pre-surgery time point. Bar plot showing Gene Set Enrichment Analysis NES for the MSigDB (Molecular Signatures Database) hallmark pathways. Significant hits (adjusted p value

References

    1. Blume-Jensen P, Hunter T. Oncogenic kinase signalling. Nature 2001;411:355–65. 10.1038/35077225
    1. Gherardi E, Birchmeier W, Birchmeier C, et al. . Targeting MET in cancer: rationale and progress. Nat Rev Cancer 2012;12:89–103. 10.1038/nrc3205
    1. Lemke G, Rothlin CV. Immunobiology of the TAM receptors. Nat Rev Immunol 2008;8:327–36. 10.1038/nri2303
    1. Paolino M, Penninger JM. The role of TAM family receptors in immune cell function: implications for cancer therapy. Cancers 2016;810.3390/cancers8100097
    1. Akalu YT, Rothlin CV, Ghosh S. TAM receptor tyrosine kinases as emerging targets of innate immune checkpoint blockade for cancer therapy. Immunol Rev 2017;276:165–77. 10.1111/imr.12522
    1. Terme M, Pernot S, Marcheteau E, et al. . VEGFA-VEGFR pathway blockade inhibits tumor-induced regulatory T-cell proliferation in colorectal cancer. Cancer Res 2013;73:539–49. 10.1158/0008-5472.CAN-12-2325
    1. Lapeyre-Prost A, Terme M, Pernot S, et al. . Immunomodulatory activity of VEGF in cancer. Int Rev Cell Mol Biol 2017;330:295–342. 10.1016/bs.ircmb.2016.09.007
    1. Sharma P, Hu-Lieskovan S, Wargo JA, et al. . Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 2017;168:707–23. 10.1016/j.cell.2017.01.017
    1. Ozao-Choy J, Ma G, Kao J, et al. . The novel role of tyrosine kinase inhibitor in the reversal of immune suppression and modulation of tumor microenvironment for immune-based cancer therapies. Cancer Res 2009;69:2514–22. 10.1158/0008-5472.CAN-08-4709
    1. Pircher A, Wolf D, Heidenreich A, et al. . Synergies of targeting tumor angiogenesis and immune checkpoints in non-small cell lung cancer and renal cell cancer: from basic concepts to clinical reality. Int J Mol Sci 2017;1810.3390/ijms18112291
    1. Rini BI, Plimack ER, Stus V, et al. . Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med 2019;380:1116–27. 10.1056/NEJMoa1816714
    1. Makker V, Taylor MH, Aghajanian C, et al. . Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer. J Clin Oncol 2020;38:2981–92. 10.1200/JCO.19.02627
    1. Du W, Huang H, Sorrelle N, et al. . Sitravatinib potentiates immune checkpoint blockade in refractory cancer models. JCI Insight 2018;310.1172/jci.insight.124184
    1. Burtness B. KEYNOTE-048: phase 3 study of first-line pembrolizumab for recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC), 2018.
    1. Ferris RL, Blumenschein G, Fayette J, et al. . Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med 2016;375:1856–67. 10.1056/NEJMoa1602252
    1. Cramer JD, Burtness B, Ferris RL. Immunotherapy for head and neck cancer: recent advances and future directions. Oral Oncol 2019;99:104460. 10.1016/j.oraloncology.2019.104460
    1. Stafford M, Kaczmar J. The neoadjuvant paradigm reinvigorated: a review of pre-surgical immunotherapy in HNSCC. Cancers Head Neck 2020;5:4. 10.1186/s41199-020-00052-8
    1. Uppaluri R, Campbell KM, Egloff AM. Neoadjuvant and adjuvant pembrolizumab in resectable locally advanced, human Papillomavirus-Unrelated head and neck cancer: a multicenter, phase 2 trial. Clin Cancer Res 2020.
    1. Melero I, Berman DM, Aznar MA, et al. . Evolving synergistic combinations of targeted immunotherapies to combat cancer. Nat Rev Cancer 2015;15:457–72. 10.1038/nrc3973
    1. Sato-Kaneko F, Yao S, Ahmadi A, et al. . Combination immunotherapy with TLR agonists and checkpoint inhibitors suppresses head and neck cancer. JCI Insight 2017;210.1172/jci.insight.93397
    1. Uehara M, Sano K, Ikeda H, et al. . Expression of vascular endothelial growth factor and prognosis of oral squamous cell carcinoma. Oral Oncol 2004;40:321–5. 10.1016/j.oraloncology.2003.08.020
    1. Brand TM, Iida M, Stein AP, et al. . AXL is a logical molecular target in head and neck squamous cell carcinoma. Clin Cancer Res 2015;21:2601–12. 10.1158/1078-0432.CCR-14-2648
    1. Hartmann S, Bhola NE, Grandis JR. HGF/Met signaling in head and neck cancer: impact on the tumor microenvironment. Clin Cancer Res 2016;22:4005–13. 10.1158/1078-0432.CCR-16-0951
    1. Oliva M, Spreafico A, Taberna M, et al. . Immune biomarkers of response to immune-checkpoint inhibitors in head and neck squamous cell carcinoma. Ann Oncol 2019;30:57–67. 10.1093/annonc/mdy507
    1. Chinn SB, Myers JN. Oral cavity carcinoma: current management, controversies, and future directions. J Clin Oncol 2015;33:3269–76. 10.1200/JCO.2015.61.2929
    1. Farlow JL, Birkeland AC, Swiecicki PL, et al. . Window of opportunity trials in head and neck cancer. J Cancer Metastasis Treat 2019;510.20517/2394-4722.2018.100
    1. Dindo D, Demartines N, Clavien P-A. Classification of surgical complications: a new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg 2004;240:205–13. 10.1097/
    1. Liberzon A, Birger C, Thorvaldsdóttir H, et al. . The molecular signatures database (MSigDB) hallmark gene set collection. Cell Syst 2015;1:417–25. 10.1016/j.cels.2015.12.004
    1. Subramanian A, Tamayo P, Mootha VK, 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. 10.1073/pnas.0506580102
    1. Toustrup K, Sørensen BS, Nordsmark M, et al. . Development of a hypoxia gene expression classifier with predictive impact for hypoxic modification of radiotherapy in head and neck cancer. Cancer Res 2011;71:5923–31. 10.1158/0008-5472.CAN-11-1182
    1. Brooks JM, Menezes AN, Ibrahim M, et al. . Development and validation of a combined hypoxia and immune prognostic classifier for head and neck cancer. Clin Cancer Res 2019;25:5315–28. 10.1158/1078-0432.CCR-18-3314
    1. Schoenfeld JD, Hanna GJ, Jo VY, et al. . Neoadjuvant nivolumab or nivolumab plus ipilimumab in untreated oral cavity squamous cell carcinoma: a phase 2 open-label randomized clinical trial. JAMA Oncol 2020;6:1563–70. 10.1001/jamaoncol.2020.2955
    1. Bossi P, Lo Vullo S, Guzzo M, et al. . Preoperative chemotherapy in advanced resectable OCSCC: long-term results of a randomized phase III trial. Ann Oncol 2014;25:462–6. 10.1093/annonc/mdt555
    1. Zuur L, Vos JL, Elbers JB, et al. . LBA40 neoadjuvant nivolumab and nivolumab plus ipilimumab induce (near-) complete responses in patients with head and neck squamous cell carcinoma: the IMCISION trial. Ann Oncol 2020;31:S1169. 10.1016/j.annonc.2020.08.2270
    1. Huang S-H, O'Sullivan B. Oral cancer: current role of radiotherapy and chemotherapy. Med Oral Patol Oral Cir Bucal 2013;18:e233–40. 10.4317/medoral.18772
    1. Chen W-C, Lai C-H, Fang C-C, et al. . Identification of high-risk subgroups of patients with oral cavity cancer in need of postoperative adjuvant radiotherapy or chemo-radiotherapy. Medicine 2016;95:e3770. 10.1097/MD.0000000000003770
    1. Woody NM, Ward MC, Koyfman SA, et al. . Adjuvant chemoradiation after surgical resection in elderly patients with high-risk squamous cell carcinoma of the head and neck: a national cancer database analysis. Int J Radiat Oncol Biol Phys 2017;98:784–92. 10.1016/j.ijrobp.2017.03.019
    1. Stroun M, Lyautey J, Lederrey C, et al. . About the possible origin and mechanism of circulating DNA. Clinica Chimica Acta 2001;313:139–42. 10.1016/S0009-8981(01)00665-9
    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;165:35–44. 10.1016/j.cell.2016.02.065
    1. Tsukita Y, Fujino N, Miyauchi E, et al. . Axl kinase drives immune checkpoint and chemokine signalling pathways in lung adenocarcinomas. Mol Cancer 2019;18:24. 10.1186/s12943-019-0953-y
    1. Coombes RC, Page K, Salari R, et al. . Personalized detection of circulating tumor DNA Antedates breast cancer metastatic recurrence. Clin Cancer Res 2019;25:4255–63. 10.1158/1078-0432.CCR-18-3663
    1. Bratman SV, Yang SYC, Iafolla MAJ, et al. . Personalized circulating tumor DNA analysis as a predictive biomarker in solid tumor patients treated with pembrolizumab. Nat Cancer 2020;1:873–81. 10.1038/s43018-020-0096-5
    1. Butler A, Hoffman P, Smibert P, et al. . Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol 2018;36:411–20. 10.1038/nbt.4096
    1. Mohanraj S, Díaz-Mejía JJ, Pham MD, et al. . Crescent: cancer single cell expression toolkit. Nucleic Acids Res 2020;48:W372–9. 10.1093/nar/gkaa437
    1. Diaz-Mejia JJ, Meng EC, Pico AR, et al. . Evaluation of methods to assign cell type labels to cell clusters from single-cell RNA-sequencing data. F1000Res 2019;8:296. 10.12688/f1000research.18490.1

Source: PubMed

3
Subscribe