A pilot study of neoadjuvant combination of anti-PD-1 camrelizumab and VEGFR2 inhibitor apatinib for locally advanced resectable oral squamous cell carcinoma

Wu-Tong Ju, Rong-Hui Xia, Dong-Wang Zhu, Sheng-Jin Dou, Guo-Pei Zhu, Min-Jun Dong, Li-Zhen Wang, Qi Sun, Tong-Chao Zhao, Zhi-Hang Zhou, Si-Yuan Liang, Ying-Ying Huang, Yong Tang, Si-Cheng Wu, Jing Xia, Shi-Qing Chen, Yue-Zong Bai, Jiang Li, Qi Zhu, Lai-Ping Zhong, Wu-Tong Ju, Rong-Hui Xia, Dong-Wang Zhu, Sheng-Jin Dou, Guo-Pei Zhu, Min-Jun Dong, Li-Zhen Wang, Qi Sun, Tong-Chao Zhao, Zhi-Hang Zhou, Si-Yuan Liang, Ying-Ying Huang, Yong Tang, Si-Cheng Wu, Jing Xia, Shi-Qing Chen, Yue-Zong Bai, Jiang Li, Qi Zhu, Lai-Ping Zhong

Abstract

Novel neoadjuvant therapy regimens are warranted for oral squamous cell carcinoma (OSCC). In this phase I trial (NCT04393506), 20 patients with locally advanced resectable OSCC receive three cycles of camrelizumab (200 mg, q2w) and apatinib (250 mg, once daily) before surgery. The primary endpoints are safety and major pathological response (MPR, defined as ≤10% residual viable tumour cells). Secondary endpoints include 2-year survival rate and local recurrence rate (not reported due to inadequate follow-up). Exploratory endpoints are the relationships between PD-L1 combined positive score (CPS, defined as the number of PD-L1-stained cells divided by the total number of viable tumour cells, multiplied by 100) and other immunological and genomic biomarkers and response. Neoadjuvant treatment is well-tolerated, and the MPR rate is 40% (8/20), meeting the primary endpoint. All five patients with CPS ˃10 achieve MPR. Post-hoc analysis show 18-month locoregional recurrence and survival rates of 10.5% (95% CI: 0%-24.3%) and 95% (95% CI: 85.4%-100.0%), respectively. Patients achieving MPR show more CD4+ T-cell infiltration than those without MPR (P = 0.02), and decreased CD31 and ɑ-SMA expression levels are observed after neoadjuvant therapy. In conclusion, neoadjuvant camrelizumab and apatinib is safe and yields a promising MPR rate for OSCC.

Conflict of interest statement

J.X., S.C. and Y.B. are employees of the company 3D Medicines Inc. The remaining authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. Trial flowchart.
Fig. 1. Trial flowchart.
21 patients were enrolled in this trial; 20 patients received neoadjuvant therapy and surgery; among them, 18 patients received adjuvant therapy.
Fig. 2. Efficacy of neoadjuvant therapy.
Fig. 2. Efficacy of neoadjuvant therapy.
A Residual viable tumour cell (RVT) ratio, combined positive score (CPS), and radiographic partial response (PR) in 20 patients. The percentage of RVT was evaluated on resected tumour slides after surgery. The CPS was defined as the total number of programmed cell death-ligand 1-stained cells (including tumour cells, tumour-associated lymphocytes, and macrophages) divided by the total number of viable tumour cells plus 100. Radiographic response according to RECIST 1.1 criteria was performed on the basis of imaging examinations before and after neoadjuvant therapy (green triangles for the MPR group [n = 8], black triangles for the non-MPR group [n = 12]). B In the patient who achieved pathological complete response, images of the oral tongue (left) and magnetic resonance imaging (right) before (upper) and after (lower) neoadjuvant therapy are shown. Source data are provided as a Source Data file.
Fig. 3. Genetic and tumour-infiltrating lymphocyte analyses.
Fig. 3. Genetic and tumour-infiltrating lymphocyte analyses.
A Mutations as assessed by next-generation sequencing of baseline primary tumour samples. A column represents a patient. The percentages listed on the right represent the proportion of samples harbouring a mutation in the gene listed on the left. Bottom bars show pathological response (MPR [n = 8] or non-MPR [n = 7]) and combined positive score (˃10 [n = 5] or ≤10 [n = 10]) distribution. B Comparison of TMB in baseline tumour samples between the MPR and non-MPR groups (green dots for the MPR group [n = 7], grey dots for the non-MPR group [n = 8]). Quantitative graphs of the infiltration density of CD8+ (C) and CD4+ (D) T cells in tumour samples before and after neoadjuvant therapy (green dots for the MPR group [n = 8], grey dots for the non-MPR group [n = 12]). The significance of the differences between before and after neoadjuvant therapy was tested using a two-sided Wilcoxon signed-rank test; for differences between the MPR and non-MPR groups, the significance was tested using a two-sided Mann‒Whitney test. Bars represent the mean with SD. Source data are provided as a Source Data file.
Fig. 4. Features of the hyperprogressive disease.
Fig. 4. Features of the hyperprogressive disease.
In patient No. 14, who showed hyperprogressive disease: radiographic and H&E staining images before (A) and after (B) neoadjuvant therapy. C Multiplex immunofluorescence images of the tumour site before and after neoadjuvant therapy. Primary antibodies targeting CD163, CD68, PD-1, CD8, PD-L1, and Pan-CK were used. Nuclei acids were stained with DAPI. D Comparison of fluorescence intensity for CD8+ and CD163+ cells. After staining, slides were scanned, and multilayer images were used for quantitative image analysis. The quantities of various cell populations were expressed as the number of stained cells per square millimetre in all nucleated cells. Due to limited tumour tissue obtained by biopsy, immunofluorescence experiments were performed without repetition. Source data are provided as a Source Data file.
Fig. 5. Anti-angiogenesis evaluation.
Fig. 5. Anti-angiogenesis evaluation.
A Representative immunofluorescence staining images of before and after neoadjuvant therapy tumour sections (Case No. 7, green for CD31, red for α-SMA, blue for DAPI). B Fluorescence intensity of CD31 and α-SMA expression before and after neoadjuvant therapy tumour tissues in all 20 patients. Whiskers represent min to max. Bounds of boxes represent 25th and 75th percentiles, centres represent medians, whiskers represent min to max. The significance for differences between before- and after neoadjuvant therapy was tested using a two-sided Wilcoxon signed-rank test. C Comparison of baseline CD31 and α-SMA fluorescence intensity between the MPR and non-MPR groups (n = 8 in the MPR group, n = 12 in the non-MPR group). The significance of the differences between the MPR and non-MPR groups was tested using a two-sided Mann‒Whitney test. Bars represent the mean with SD. Source data are provided as a Source Data file.

References

    1. Johnson DE, et al. Head and neck squamous cell carcinoma. Nat. Rev. Dis. Prim. 2020;6:92. doi: 10.1038/s41572-020-00224-3.
    1. Specenier P, Vermorken JB. Optimizing treatments for recurrent or metastatic head and neck squamous cell carcinoma. Expert Rev. Anticancer Ther. 2018;18:901–915. doi: 10.1080/14737140.2018.1493925.
    1. Funt SA, Chapman PB. The role of neoadjuvant trials in drug development for solid tumors. Clin. Cancer Res. 2016;22:2323–2328. doi: 10.1158/1078-0432.CCR-15-1961.
    1. Zhong LP, et al. Randomized phase III trial of induction chemotherapy with docetaxel, cisplatin, and fluorouracil followed by surgery versus up-front surgery in locally advanced resectable oral squamous cell carcinoma. J. Clin. Oncol. 2013;31:744–751. doi: 10.1200/JCO.2012.43.8820.
    1. Bossi P, et al. Preoperative chemotherapy in advanced resectable OCSCC: long-term results of a randomized phase III trial. Ann. Oncol. 2014;25:462–466. doi: 10.1093/annonc/mdt555.
    1. Seiwert TY, 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. doi: 10.1016/S1470-2045(16)30066-3.
    1. Ferris RL, et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N. Engl. J. Med. 2016;375:1856–1867. doi: 10.1056/NEJMoa1602252.
    1. Liu J, et al. Improved efficacy of neoadjuvant compared to adjuvant immunotherapy to eradicate metastatic disease. Cancer Discov. 2016;6:1382–1399. doi: 10.1158/-16-0577.
    1. Chalabi M, et al. Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat. Med. 2020;26:566–576. doi: 10.1038/s41591-020-0805-8.
    1. Huang AC, et al. A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat. Med. 2019;25:454–461. doi: 10.1038/s41591-019-0357-y.
    1. Cloughesy TF, et al. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat. Med. 2019;25:477–486. doi: 10.1038/s41591-018-0337-7.
    1. Forde PM, et al. Neoadjuvant PD-1 blockade in resectable lung cancer. N. Engl. J. Med. 2018;378:1976–1986. doi: 10.1056/NEJMoa1716078.
    1. Schoenfeld JD, 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;6:1563–1570. doi: 10.1001/jamaoncol.2020.2955.
    1. Wise-Draper T, et al. 809 phase 2 trial of neoadjuvant and adjuvant PD-1 checkpoint blockade in local-regionally advanced, resectable HNSCC indicates pathological response is associated with high disease-free survival. J. Immunother. Cancer. 2020;8:A484–A485.
    1. Li Q, et al. Low-dose anti-angiogenic therapy sensitizes breast cancer to PD-1 blockade. Clin. Cancer Res. 2020;26:1712–1724. doi: 10.1158/1078-0432.CCR-19-2179.
    1. Fukumura D, Kloepper J, Amoozgar Z, Duda DG, Jain RK. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat. Rev. Clin. Oncol. 2018;15:325–340. doi: 10.1038/nrclinonc.2018.29.
    1. Zaremba A, et al. The concepts of rechallenge and retreatment with immune checkpoint blockade in melanoma patients. Eur. J. Cancer. 2021;155:268–280. doi: 10.1016/j.ejca.2021.07.002.
    1. Saeed A, Park R, Sun W. The integration of immune checkpoint inhibitors with VEGF targeted agents in advanced gastric and gastroesophageal adenocarcinoma: a review on the rationale and results of early phase trials. J. Hematol. Oncol. 2021;14:13. doi: 10.1186/s13045-021-01034-0.
    1. Zhang B, et al. Phase II clinical trial using camrelizumab combined with apatinib and chemotherapy as the first-line treatment of advanced esophageal squamous cell carcinoma. Cancer Commun. 2020;40:711–720. doi: 10.1002/cac2.12119.
    1. Lan C, et al. Camrelizumab plus apatinib in patients with advanced cervical cancer (CLAP): a multicenter, open-label, single-arm, phase II trial. J. Clin. Oncol. 2020;38:4095–4106. doi: 10.1200/JCO.20.01920.
    1. Xu J, et al. Camrelizumab in combination with apatinib in patients with advanced hepatocellular carcinoma (RESCUE): a nonrandomized, open-label, phase II trial. Clin. Cancer Res. 2021;27:1003–1011. doi: 10.1158/1078-0432.CCR-20-2571.
    1. Xu J, et al. Anti-PD-1 antibody SHR-1210 combined with apatinib for advanced hepatocellular carcinoma, gastric, or esophagogastric junction cancer: an open-label, dose escalation and expansion study. Clin. Cancer Res. 2019;25:515–523. doi: 10.1158/1078-0432.CCR-18-2484.
    1. Liu, J. et al. Efficacy and safety of camrelizumab combined with apatinib in advanced triple-negative breast cancer: an open-label phase II trial. J. Immunother. Cancer8, e000696 (2020).
    1. Inhestern J, et al. A two-arm multicenter phase II trial of one cycle chemoselection split-dose docetaxel, cisplatin and 5-fluorouracil (TPF) induction chemotherapy before two cycles of split TPF followed by curative surgery combined with postoperative radiotherapy in patients with locally advanced oral and oropharyngeal squamous cell cancer (TISOC-1) Ann. Oncol. 2017;28:1917–1922. doi: 10.1093/annonc/mdx202.
    1. Licitra L, et al. Primary chemotherapy in resectable oral cavity squamous cell cancer: a randomized controlled trial. J. Clin. Oncol. 2003;21:327–333. doi: 10.1200/JCO.2003.06.146.
    1. Yen CJ, et al. Sequential therapy of neoadjuvant biochemotherapy with cetuximab, paclitaxel, and cisplatin followed by cetuximab-based concurrent bioradiotherapy in high-risk locally advanced oral squamous cell carcinoma: final analysis of a phase 2 clinical trial. Head. Neck. 2019;41:1703–1712. doi: 10.1002/hed.25640.
    1. Uppaluri R, et al. Biomarker and tumor responses of oral cavity squamous cell carcinoma to trametinib: a phase II neoadjuvant window-of-opportunity clinical trial. Clin. Cancer Res. 2017;23:2186–2194. doi: 10.1158/1078-0432.CCR-16-1469.
    1. Duhen R, et al. Neoadjuvant anti-OX40 (MEDI6469) therapy in patients with head and neck squamous cell carcinoma activates and expands antigen-specific tumor-infiltrating T cells. Nat. Commun. 2021;12:1047. doi: 10.1038/s41467-021-21383-1.
    1. Uppaluri R, 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;26:5140–5152. doi: 10.1158/1078-0432.CCR-20-1695.
    1. Tewari KS, et al. Bevacizumab for advanced cervical cancer: final overall survival and adverse event analysis of a randomised, controlled, open-label, phase 3 trial (Gynecologic Oncology Group 240) Lancet. 2017;390:1654–1663. doi: 10.1016/S0140-6736(17)31607-0.
    1. Zheng Y, et al. Effect of apatinib plus neoadjuvant chemotherapy followed by resection on pathologic response in patients with locally advanced gastric adenocarcinoma: a single-arm, open-label, phase II trial. Eur. J. Cancer. 2020;130:12–19. doi: 10.1016/j.ejca.2020.02.013.
    1. Ferris, R. L. et al. Neoadjuvant nivolumab for patients with resectable HPV-positive and HPV-negative squamous cell carcinomas of the head and neck in the CheckMate 358 trial. J. Immunother. Cancer9, e002568 (2021).
    1. Hecht, M. et al. Safety and efficacy of single cycle induction treatment with cisplatin/docetaxel/ durvalumab/tremelimumab in locally advanced HNSCC: first results of CheckRad-CD8. J. Immunother. Cancer8, e001378 (2020).
    1. Leidner, R. et al. Neoadjuvant immunoradiotherapy results in high rate of complete pathological response and clinical to pathological downstaging in locally advanced head and neck squamous cell carcinoma. J. Immunother. Cancer9, e002485 (2021).
    1. Borel, C., Jung, A. C. & Burgy, M. Immunotherapy breakthroughs in the treatment of recurrent or metastatic head and neck squamous cell carcinoma. Cancers12, 2691 (2020).
    1. Tetzlaff MT, et al. Pathological assessment of resection specimens after neoadjuvant therapy for metastatic melanoma. Ann. Oncol. 2018;29:1861–1868. doi: 10.1093/annonc/mdy226.
    1. Cottrell TR, et al. Pathologic features of response to neoadjuvant anti-PD-1 in resected non-small-cell lung carcinoma: a proposal for quantitative immune-related pathologic response criteria (irPRC) Ann. Oncol. 2018;29:1853–1860. doi: 10.1093/annonc/mdy218.
    1. Merlino DJ, et al. Discordant responses between primary head and neck tumors and nodal metastases treated with neoadjuvant nivolumab: correlation of radiographic and pathologic treatment effect. Front. Oncol. 2020;10:566315. doi: 10.3389/fonc.2020.566315.
    1. Seymour L, et al. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017;18:e143–e152. doi: 10.1016/S1470-2045(17)30074-8.
    1. Provencio M, et al. Neoadjuvant chemotherapy and nivolumab in resectable non-small-cell lung cancer (NADIM): an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol. 2020;21:1413–1422. doi: 10.1016/S1470-2045(20)30453-8.
    1. Cancer Genome Atlas Network. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 2015;517:576–582. doi: 10.1038/nature14129.
    1. Vos JL, et al. Neoadjuvant immunotherapy with nivolumab and ipilimumab induces major pathological responses in patients with head and neck squamous cell carcinoma. Nat. Commun. 2021;12:7348. doi: 10.1038/s41467-021-26472-9.
    1. Parra ER, et al. Effect of neoadjuvant chemotherapy on the immune microenvironment in non-small cell lung carcinomas as determined by multiplex immunofluorescence and image analysis approaches. J. Immunother. Cancer. 2018;6:48. doi: 10.1186/s40425-018-0368-0.
    1. Kim SR, et al. The implications of clinical risk factors, CAR index, and compositional changes of immune cells on hyperprogressive disease in non-small cell lung cancer patients receiving immunotherapy. BMC Cancer. 2021;21:19. doi: 10.1186/s12885-020-07727-y.
    1. Ragusa S, et al. Antiangiogenic immunotherapy suppresses desmoplastic and chemoresistant intestinal tumors in mice. J. Clin. Invest. 2020;130:1199–1216. doi: 10.1172/JCI129558.
    1. Mancuso MR, et al. Rapid vascular regrowth in tumors after reversal of VEGF inhibition. J. Clin. Invest. 2006;116:2610–2621. doi: 10.1172/JCI24612.
    1. Tian L, et al. Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming. Nature. 2017;544:250–254. doi: 10.1038/nature21724.
    1. Song Y, et al. Anti-angiogenic agents in combination with immune checkpoint inhibitors: a promising strategy for cancer treatment. Front. Immunol. 2020;11:1956. doi: 10.3389/fimmu.2020.01956.
    1. Huang Y, et al. Improving immune-vascular crosstalk for cancer immunotherapy. Nat. Rev. Immunol. 2018;18:195–203. doi: 10.1038/nri.2017.145.
    1. Lee WS, Yang H, Chon HJ, Kim C. Combination of anti-angiogenic therapy and immune checkpoint blockade normalizes vascular-immune crosstalk to potentiate cancer immunity. Exp. Mol. Med. 2020;52:1475–1485. doi: 10.1038/s12276-020-00500-y.
    1. Pradhan M, Chocry M, Gibbons DL, Sepesi B, Cascone T. Emerging biomarkers for neoadjuvant immune checkpoint inhibitors in operable non-small cell lung cancer. Transl. Lung Cancer Res. 2021;10:590–606. doi: 10.21037/tlcr-20-573.
    1. Pataer A, et al. Histopathologic response criteria predict survival of patients with resected lung cancer after neoadjuvant chemotherapy. J. Thorac. Oncol. 2012;7:825–832. doi: 10.1097/JTO.0b013e318247504a.
    1. Brahmer JR, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2018;36:1714–1768. doi: 10.1200/JCO.2017.77.6385.
    1. Clavien PA, et al. The Clavien-Dindo classification of surgical complications: five-year experience. Ann. Surg. 2009;250:187–196. doi: 10.1097/SLA.0b013e3181b13ca2.

Source: PubMed

3
Prenumerera