Anlotinib Combined with Toripalimab as Second-Line Therapy for Advanced, Relapsed Gastric or Gastroesophageal Junction Carcinoma

Man Jiang, Chuantao Zhang, Yabin Hu, Tianjun Li, Guangjie Yang, Guanqun Wang, Jingjuan Zhu, Changfeng Shao, Helei Hou, Na Zhou, Kewei Liu, Xiaochun Zhang, Man Jiang, Chuantao Zhang, Yabin Hu, Tianjun Li, Guangjie Yang, Guanqun Wang, Jingjuan Zhu, Changfeng Shao, Helei Hou, Na Zhou, Kewei Liu, Xiaochun Zhang

Abstract

Our study aimed to explore the efficacy and safety of anlotinib-toripalimab combination therapy as a second-line treatment for advanced relapsed gastric or gastroesophageal junction carcinoma (GC/GEJC). In this single arm, single-center extension clinical trial, patients with advanced relapsed GC/GEJC received toripalimab (240 mg, intravenously over 60 minutes, once every 2 weeks) plus anlotinib (12 mg/day, orally, 2 weeks on and 1 week off, every 3 weeks) as second-line therapy. There were 29 patients who achieved partial response, and the ORR was 32.3% (95% CI, 26.6%-38.5%). Grade 3 treatment-related adverse events (TRAEs) were recorded in 7 participants (11.3%), all of which were manageable. The PFS and OS were 4.0 and 11.1 months, respectively. Patients with programmed death-ligand 1 (PD-L1) positive expression showed numerically longer OS than the negative ones although the difference was not significantly. The tumor mutational burden-high (TMB-H) group showed a significantly better OS (P = .05) than the TMB-Low (TMB-L) group. Next-generation sequencing (NGS) revealed that fibroblast growth factor receptor 2 (FGFR2) mutations positively correlated with target lesion reduction (odds ratio [OR] = 0.14; P = .02). The new regimen increased tumor-infiltration of CD8+ T and CD3+ T cells. Furthermore, a patient-derived organoid (PDO) study indicated that anlotinib could promote an immune-supportive tumor microenvironment. As conclusion, the anlotinib-toripalimab combination showed promising efficacy and favorable safety as a second-line treatment for advanced, relapsed GC/GEJC. The PD-L1 expression, TMB, and FGFR2 mutation are potential biomarkers for predicting the efficacy of this regimen (ClinicalTrials.gov registration number: NCT04713059).

Keywords: anlotinib; gastric or gastroesophageal junction carcinoma; second-line therapy; toripalimab; tumor microenviroment.

© The Author(s) 2022. Published by Oxford University Press.

Figures

Figure 1.
Figure 1.
Efficacy analysis. (1) Waterfall plot of best percentage change from baseline in size of target tumor lesion; (2) Percentage change in lesion diameters over time; (3) Kaplan-Meier estimates of progression-free survival (PFS); (4) Kaplan-Meier estimates of overall survival (OS).
Figure 2.
Figure 2.
(1) ORR (A), PFS (B) and OS (C) of PD-L1 negative or positive patients; (2) ORR (A), PFS (B) and OS (C) of TMB-H, TMB-L and TMB-H (excluding MSI-H); (3) Gene mutation analysis of all the enrolled patients; (4) The ORR of FGFR2-mutated patients, and the FGFR2 mutation ratio in PD-L1 positive or TMB-H groups (A), PFS (B) and OS (C) of FGFR2-mutated patients.
Figure 2.
Figure 2.
(1) ORR (A), PFS (B) and OS (C) of PD-L1 negative or positive patients; (2) ORR (A), PFS (B) and OS (C) of TMB-H, TMB-L and TMB-H (excluding MSI-H); (3) Gene mutation analysis of all the enrolled patients; (4) The ORR of FGFR2-mutated patients, and the FGFR2 mutation ratio in PD-L1 positive or TMB-H groups (A), PFS (B) and OS (C) of FGFR2-mutated patients.
Figure 3.
Figure 3.
Tumor microenvironment analysis of patient GC-20 who was underwent surgery after 8 courses of combination treatment: (1) The immune microenvironment of the tumor before combination treatment: 1) the immune microenvironment panorama, 2) expression of CD3 and CD8, 3) expression of PD-1, 4) expression of PD-L1, 5) expression of CD8, 6) expression of CD3; (2) The immune microenvironment of the tumor after combination treatment: 1) the immune microenvironment panorama, 2) expression of CD3 and CD8, 3) expression of PD-1, 4) expression of PD-L1, 5) expression of CD8, 6) expression of CD3 (CD8: red; CD3: cyan; PD-L1: yellow; PD-1: green; panCK: purple; DAPI: blue); (3) PET-CT scan of patient GC-20 before and after the combination therapy: 1) the PET-CT showed an uneven thickening of uneven thickening of the gastric body wall with (SUVmax:3.2) before treatment. After treatment, the lateral wall of the greater curvature of the stomach was thickened (SUVmax 3.6), and the lesion was red uced; 2) the PET-CT showed multiple enlarged lymph nodes around left of the gastric artery, great curvature of the gastric body, right lower part of the gastric antrum, upper part of the pancreas, and around the abdominal aorta (SUVmax: 5.1) before the treatment. After the treatment, there was no abnormal metabolism or decreased metabolism in the retroperitoneum; 3) the PET-CT showed multiple lymph nodes could be observed in left subphrenic fat, the anterior omentum of the transverse colon and the left pelvic descending colon (SUVmax: 3.8). After treatment, multiple pelvic lymph nodes in the left anterior omentum of transverse colon could not been observed.
Figure 3.
Figure 3.
Tumor microenvironment analysis of patient GC-20 who was underwent surgery after 8 courses of combination treatment: (1) The immune microenvironment of the tumor before combination treatment: 1) the immune microenvironment panorama, 2) expression of CD3 and CD8, 3) expression of PD-1, 4) expression of PD-L1, 5) expression of CD8, 6) expression of CD3; (2) The immune microenvironment of the tumor after combination treatment: 1) the immune microenvironment panorama, 2) expression of CD3 and CD8, 3) expression of PD-1, 4) expression of PD-L1, 5) expression of CD8, 6) expression of CD3 (CD8: red; CD3: cyan; PD-L1: yellow; PD-1: green; panCK: purple; DAPI: blue); (3) PET-CT scan of patient GC-20 before and after the combination therapy: 1) the PET-CT showed an uneven thickening of uneven thickening of the gastric body wall with (SUVmax:3.2) before treatment. After treatment, the lateral wall of the greater curvature of the stomach was thickened (SUVmax 3.6), and the lesion was red uced; 2) the PET-CT showed multiple enlarged lymph nodes around left of the gastric artery, great curvature of the gastric body, right lower part of the gastric antrum, upper part of the pancreas, and around the abdominal aorta (SUVmax: 5.1) before the treatment. After the treatment, there was no abnormal metabolism or decreased metabolism in the retroperitoneum; 3) the PET-CT showed multiple lymph nodes could be observed in left subphrenic fat, the anterior omentum of the transverse colon and the left pelvic descending colon (SUVmax: 3.8). After treatment, multiple pelvic lymph nodes in the left anterior omentum of transverse colon could not been observed.
Figure 4.
Figure 4.
PDO studies. (1) HE staining of the patients tumor tissue; (2) HE staining of the patient-derived gastric cancer organoid (PDO); (3) the direct efficacy of anlotinib on the PDOs; (4) viability assay of the different treatment tumor cells from the PDOs cultivated for 7 days; (5) STRING analysis to study the potential mechanism; (6) western-blot analysis of the expression of STAT1 and STAT3 after anlotinib treatment.
Figure 4.
Figure 4.
PDO studies. (1) HE staining of the patients tumor tissue; (2) HE staining of the patient-derived gastric cancer organoid (PDO); (3) the direct efficacy of anlotinib on the PDOs; (4) viability assay of the different treatment tumor cells from the PDOs cultivated for 7 days; (5) STRING analysis to study the potential mechanism; (6) western-blot analysis of the expression of STAT1 and STAT3 after anlotinib treatment.

References

    1. Bray F, Ferlay J, Soerjomataram I, et al. . Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018;68(6):394-424. 10.3322/caac.21492.
    1. Ueno M, Doi A, Sunami T, et al. . Delivery rate of patients with advanced gastric cancer to third-line chemotherapy and those patients’ characteristics: an analysis in real-world setting. J Gastrointest Oncol 2019;10(5):957-957. 10.21037/jgo.2019.05.07.
    1. Gómez-Ulloa D, Amonkar M, Kothari S, et al. . Real-world treatment patterns, healthcare resource use and clinical outcomes of patients receiving second line therapy for advanced or metastatic gastric cancer. BMC Gastroenterol. 2020;20(1):133. 10.1186/s12876-020-01232-z.
    1. Chung HC, Bang YJ, Fuchs C S, et al. . First-line pembrolizumab/placebo plus trastuzumab and chemotherapy in HER2-positive advanced gastric cancer: KEYNOTE-811. Future Oncol. 2021;17(5):491-410. 10.2217/fon-2020-0737.
    1. Janjigian YY, Shitara K, Moehler M, 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;S0140-6736(21):00797-00792. 10.1016/S0140-6736(21)00797-2.
    1. Siu LL, Ivy SP, Dixon EL, et al. . Challenges and opportunities in adapting clinical trial design for immunotherapies. Clin Cancer Res. 2017;23(17):4950-4958. 10.1158/1078-0432.CCR-16-3079 .
    1. Tang J, Shalabi A, Hubbard-Lucey VM.. Comprehensive analysis of the clinical immuno-oncology landscape. Ann Oncol. 2018;29(1):84-91. 10.1093/annonc/mdx755.
    1. Fukuoka S, Hara H, Takahashi N, et al. . Regorafenib plus nivolumab in patients with advanced gastric or colorectal cancer: an open-label, dose-escalation, and dose-expansion phase Ib trial (REGONIVO, EPOC1603). J Clin Oncol. 2020;38(18):2053-2061. 10.1200/JCO.19.03296 . Epub 2020 Apr 28.
    1. Chen H, Jiang T, Lin F, et al. . PD-1 inhibitor combined with apatinib modulate the tumor microenvironment and potentiate anti-tumor effect in mice bearing gastric cancer. Int Immunopharmacol. 2021;99:107929. 10.1016/j.intimp.2021.107929.
    1. Kawazoe A, Fukuoka S, Nakamura Y, et al. . Lenvatinib plus pembrolizumab in patients with advanced gastric cancer in the first-line or second-line setting (EPOC1706): an open-label, single-arm, phase 2 trial. Lancet Oncol. 2020;21(8):1057-1065. 10.1016/S1470-2045(20)30271-0.
    1. Xu J, Zhang Y, Jia R, 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(2):515-523. 10.1158/1078-0432.CCR-18-2484 .
    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(5):325-325. 10.1038/nrclinonc.2018.29.
    1. Yi M, Jiao D, Qin S, et al. . Synergistic effect of immune checkpoint blockade and anti-angiogenesis in cancer treatment. Mol Cancer. 2019;18(1):60. 10.1186/s12943-019-0974-6.
    1. Shen G, Zheng F, Ren D, et al. . Anlotinib: a novel multi-targeting tyrosine kinase inhibitor in clinical development. J Hematol Oncol 2018;11(1):120. 10.1186/s13045-018-0664-7.
    1. Lin B, Song X, Yang D, et al. . Anlotinib inhibits angiogenesis via suppressing the activation of VEGFR2, PDGFRβ and FGFR1. Gene 2018;654:77-86. 10.1016/j.gene.2018.02.026.
    1. Syed YY. Anlotinib: first global approval. Drugs. 2018;78(10):1057-1062. 10.1007/s40265-018-0939-x.
    1. Sun Y, Niu W, Du F, et al. . Safety, pharmacokinetics, and antitumor properties of anlotinib, an oral multi-target tyrosine kinase inhibitor, in patients with advanced refractory solid tumors. J Hematol Oncol 2016;9(1):105. 10.1186/s13045-016-0332-8.
    1. Yuan M, Zhu Z, Mao W, et al. . Anlotinib combined with anti-PD-1 antibodies therapy in patients with advanced refractory solid tumors: a single-center, observational, prospective study. Front Oncol. 2021;11:683502. 10.3389/fonc.2021.683502.
    1. Fu J, Wang F, Dong LH, et al. . Preclinical evaluation of the efficacy, pharmacokinetics and immunogenicity of JS-001, a programmed cell death protein-1 (PD-1) monoclonal antibody. Acta Pharmacol Sin. 2017;38(5):710-718. 10.1038/aps.2016.161.
    1. Wei XL, Ren C, Wang FH, et al. . A phase I study of toripalimab, an anti-PD-1 antibody, in patients with refractory malignant solid tumors. Cancer Commun (Lond) 2020;40(8):345-354. 10.1002/cac2.12068.
    1. Folprecht G. Tumor mutational burden as a new biomarker for PD-1 antibody treatment in gastric cancer. Cancer Commun (Lond) 2019;39(1):74. 10.1186/s40880-019-0417-1.
    1. Wang F, Wei XL, Wang FH, et al. . Safety, efficacy and tumor mutational burden as a biomarker of overall survival benefit in chemo-refractory gastric cancer treated with toripalimab, a PD-1 antibody in phase Ib/II clinical trial NCT02915432. Ann Oncol. 2019;30(9):1479-1486. 10.1093/annonc/mdz197.
    1. Kang YK, Boku N, Satoh T, et al. . Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;390(10111):2461-2471. 10.1016/S0140-6736(17)31827-5.
    1. Fuchs CS, Doi T, Jang RW, et al. . Safety and efficacy of pembrolizumab monotherapy in patients with previously treated advanced gastric and gastroesophageal junction cancer: phase 2 clinical KEYNOTE-059 trial. JAMA Oncol 2018;4(5):e180013. 10.1001/jamaoncol.2018.0013 .
    1. Abate F, Ambrosio MR, Mundo L, et al. . Disctinct viral and mutational spectrum of endemic Burkitt lymphoma. PLoS Pathog. 2015;11:e10051581-e10051521. 10.1371/journal.ppat.1005158.
    1. Park Y, Koh J, Na HY, et al. . PD-L1 testing in gastric cancer by the combined positive score of the 22C3 PharmDx and SP263 assay with clinically relevant cut-offs. Cancer Res Treat 2020;52(3):661-670. 10.4143/crt.2019.718.
    1. Frampton GM, Fichtenholtz A, Otto GA, et al. . Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. 2013;31(11):1023-1031. 10.1038/nbt.2696.
    1. Ma C, Patel K, Singhi AD, et al. . Programmed death-ligand 1 expression is common in gastric cancer associated with epstein-barr virus or microsatellite instability. Am J Surg Pathol. 2016;40(11):1496-1410. 10.1097/PAS.0000000000000698.
    1. van de Wetering M, Francies HE, Francis JM, et al. . Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell 2015;161(4):933-912. 10.1016/j.cell.2015.03.053.
    1. Szklarczyk D, Franceschini A, Kuhn M, et al. . The STRING database in 2011: functional interaction networks of proteins, globally integrated and scored. Nucleic Acids Res. 2011;39(Database issue):D561-D568. 10.1093/nar/gkq973 .
    1. Li JJ, Tong X.. Statistical hypothesis testing versus machine learning binary classification: distinctions and guidelines. Patterns (N Y) 2020;1(7):100115. 10.1016/j.patter.2020.100115.
    1. Freites-Martinez A, Santana N, Arias-Santiago S, Viera A.. Using the Common Terminology Criteria for Adverse Events (CTCAE - Version 5.0) to evaluate the severity of adverse events of anticancer therapies. ActasDermosifiliogr (Engl Ed) 2021;112(1):90-92. 10.1016/j.ad.2019.05.009.
    1. Zhang Z, Xie T, Zhang X, et al. . Immune checkpoint inhibitors for treatment of advanced gastric or gastroesophageal junction cancer: current evidence and future perspectives. Chin J Cancer Res 2020;32(3):287-302. 10.21147/j.issn.1000-9604.2020.03.02.
    1. Samstein RM, Lee CH, Shoushtari AN, et al. . Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51:202-206. 10.1038/s41588-018-0312-8.
    1. Cristescu R, Mogg R, Ayers M, et al. . Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 2018;362:aar3593. 10.1126/science.aar3593.
    1. Goodman AM, Kato S, Bazhenova L, et al. . Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 2017;16(2):598-510. 10.1158/1535-7163.MCT-17-0386.
    1. Marabelle A, Fakih M, Lopez J, et al. . Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 2020;21(10):1353-1365. 10.1016/S1470-2045(20)30445-9.
    1. Greally M, Chou JF, Chatila WK, et al. . Clinical and molecular predictors of response to immune checkpoint inhibitors in patients with advanced esophagogastric cancer. Clin Cancer Res. 2019;25(20):6160-6169. 10.1158/1078-0432.CCR-18-3603 .
    1. Li P, Huang T, Zou Q, et al. . FGFR2 promotes expression of PD-L1 in colorectal cancer via the JAK/STAT3 signaling pathway. J Immunol. 2019;202(10):3065-3075. 10.4049/jimmunol.1801199.

Source: PubMed

3
Suscribir