Response to the rechallenge of combination immunotherapy in a patient with late-stage gastric cancer: case report
Case Report

Response to the rechallenge of combination immunotherapy in a patient with late-stage gastric cancer: case report

Zhening Zhang, Siyuan Cheng, Changsong Qi, Xiaotian Zhang, Zhi Peng, Lin Shen

Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China

Correspondence to: Zhi Peng. No.52, Fucheng Road, Haidian District, Beijing 100142, China. Email: zhipeng3@hotmail.com.

Abstract: Until present, late-stage gastric cancer (GC) remains a refractory disease featured with poor prognosis. However, the upcoming era of immunotherapy turns situations around. Programmed death-1 receptor (PD-1)/programmed death-ligand 1 (PD-L1) signaling blockade, an option of immune checkpoint inhibitor (ICI), has gained approval for the third-line treatment of patients with locally advanced and/or metastatic GC. Despite its efficacy in a few tumor subtypes, most patients are insensitive to anti-PD-1 monotherapy. Therefore, growing interests arise in combination immunotherapy. To explore the maximal potentials of ICI, oncologists also focus on ICI rechallenge. Here we present a patient with unresected stage IV, Epstein-Barr virus (EBV) unrelated, microsatellite stable and HER2 heterogeneously expressed gastric adenocarcinoma who experienced the failures of previous immunotherapy. He was then rechallenged with identical ICI regime combined with tyrosine kinase inhibiting and anti-HER2 therapy. The patient responded and tolerated well to this regimen and survived with fair living quality after 28 months of treatment. This case suggests that patients who fail frontline immunotherapy still possibly benefit from rechallenge of the same anti-PD-1 regimen. Besides, combination of ICI and other target agents might intrigue a synergistic effect, resulting in enhanced anti-tumor effect, which provides a meaningful therapeutic strategy. Relevant studies are underway of investigation and might be a hotspot for future research.

Keywords: Gastric cancer (GC); immunotherapy; immune checkpoint inhibitor (ICI); rechallenge; combination therapy; case report


Submitted Jan 12, 2021. Accepted for publication May 06, 2021.

doi: 10.21037/apm-21-83


Introduction

Gastric cancer (GC) is the fifth most common cancer with over 1,000,000 new cases annually, and also the third leading cause of cancer mortality worldwide (1). Despite that interventional endoscopic therapy or surgeries make early-stage GC curable, many late-stage patients have already been subject to local advancement and/or metastasis (stage IV) at diagnosis, losing chances of radical therapy (2,3). Traditionally, multiline sequential chemotherapy is the standard treatment principle for unresectable late-stage GC patients (4).

The marvelous discovery of immune checkpoints has revolutionized the field of cancer therapeutics. Programmed death-1 receptor (PD-1) and its ligands (PD-L1/PD-L2) are a pair of immunosuppressive molecules whose interactions downregulate T cell function. Previous studies have shed light on the important roles of PD-1/PD-L1 signaling during tumorigenesis (5). Immune checkpoint inhibitor (ICI) could efficaciously rescue anergic immune cells and restart the host anti-tumor immune response. Pembrolizumab (Keytruda) and nivolumab (Opdivo) are two anti-PD-1 monoclonal antibodies which have gained approval for the third-line treatment of locally advanced and/or metastatic GC patients due to the encouraging results of Keynote-059 and Attraction-2 (2,3). Current evidence indicated that Epstein-Barr virus (EBV)-related, mismatch repair (MMR) deficient or PD-L1 highly expressed GC have better response to immunotherapy in contrast to other subtypes in terms of overall response rate (ORR), progression-free survival (PFS) and overall survival (OS) (6,7). However, these populations only make up a small portion of late-stage GC patients. Pembrolizumab monotherapy failed to improve OS compared with optimal chemotherapy in the frontline therapy for most patients, according to the results of Keynote-061 and Keynote-062 (8,9). In that case, many questions remain undefined. First, how to expand applicable populations of ICI? Second, what are subsequent treatment options after the initial failure of ICI monotherapy? During recent years, the strategy of combination immunotherapy has been put forward, providing options to break the current dilemmas. The efficacy of combination immunotherapy in GC patients should be examined. Besides, though ICI rechallenge has been reported in other cancer types, it has never been discussed in the field of GC among past literature.

Herein, we reported the case of a male patient who developed stage IV gastric adenocarcinoma, got progressed on immunotherapy and was then rechallenged with the combinations of pembrolizumab, anlotinib, trastuzumab and ultimately attained a continuous response. We present the following case in accordance with the CARE reporting checklist (available at https://apm.amegroups.com/article/view/10.21037/apm-21-83/rc).


Case presentation

A 60-year-old male patient presented in April 2017 with a liver mass during a routine medical examination. When admitted into our hospital, he was asymptomatic except for inconspicuous body weight loss. His past medical history was uneventful. The patient completed a liver mass biopsy and the result suggested a malignant gastrointestinal origin. The patient then undertook gastroscopy and a positron emission tomography-computed tomography (PET-CT) scan, which indicated enhanced metabolism in the lesser curvature of the stomach. According to the imaging and histological evidence, he was diagnosed with moderately differentiated gastric adenocarcinoma with isolated liver metastasis (Figure 1). The clinical and molecular information of this patient is summarized in Table 1.

Figure 1 Pathologic results of the liver mass biopsy. (A) The low-medium differentiated adenocarcinoma, in which most glands were poorly structured. (H&E staining, original magnification was ×100). (B) The biopsy indicated that it was a metastasis coming from the stomach based on morphological and molecular evidence. (H&E staining, original magnification was ×100). (C) Negative PD-L1 staining in tumor cells. (IHC staining, original magnification ×100). (D) Negative HER2 amplification detected by FISH. PD-L1, programmed death ligand-1; FISH, fluorescence in situ hybridization.

Table 1

Clinical and molecular information of the patient

Items Detection method State/value
Patient-related
   PS 1
   HP infection 13C-UBT +
Tumor-related (primary tumor)
   Laurén classification H&E staining Intestinal type
   TMB NGS 12.2 mutations/Mb
   MSI status NGS MSS
   EBER expression IHC
   PD-L1 expression IHC 10% positive in tumor cells
   EGFR expression IHC +
   HER2 expression IHC ++
FISH

PS, performance status score; HP, helicobacter pylori; 13C-UBT, 13C-urea breath test; H&E, Hematoxylin and Eosin; TMB, tumor mutation burden; NGS, next-generation sequencing; MSI, microsatellite instability; MSS, microsatellite stable; EBER, EBV-encoded RNA; IHC, immunohistochemistry; PD-L1, programmed death ligand-1; EGFR, epidermal growth factor receptor; HER2, human epidermal growth factor receptor 2; FISH, fluorescence in situ hybridization.

He initially received 6 cycles of XELOX (oxaliplatin 200 mg day 1, capecitabine 1,500 mg bid day 1–14) and trastuzumab (Herceptin 440 mg day 1) since June 2017. According to the response evaluation criteria in solid tumors (RECIST 1.1), the patient was evaluated as radiologic stable disease (SD), SD and progressive disease (PD) after 2, 4 and 6 cycles of treatment, respectively. He discontinued therapies and got discharged. 2 months later, he was enrolled in the trial A Phase Ia/Ib Study of CS1001 in Subjects with Advanced Solid Tumors (NCT03312842) and initiated 2 cycles of anti-PD-L1 therapy (CS1001, 610 mg day 1). Nevertheless, he was soon out of the trial due to severe gastrointestinal bleeding. 4 weeks later, the patient was re-admitted and received S-1 (60 mg bid day 1–5, 8–12, 15–19) and synchronous irradiation of the gastric lesion (95% PGTV 50 Gy/95% PTV, 45 Gy/25 f). Five months later, a restaging computed tomography (CT) scan indicated enlarged peri-gastric lymph nodes which demonstrated PD. He accordingly adjusted the regimen to paclitaxel (270 mg day 1) and S-1 (60 mg bid day 1–14). Two cycles later, he was evaluated as SD but developed grade 3 neuropathy which impaired his life quality. Therefore, docetaxel (100 mg day 1) was commenced in place of paclitaxel. He was evaluated as SD following 4 cycles of treatment. During cycle 5, he developed grade 2 diarrhea and then discontinued docetaxel. One week later, the restaging CT revealed new-onset nodules in the retroperitoneum, pelvic cavity, residual liver and lung, which demonstrated PD. The patient then changed regimen to irinotecan (249 mg day 1). One week later, the chest CT scan revealed an enlarged pulmonary nodule, and the abdominal and pelvic CT scan demonstrated an increased density of peritoneum and omentum majus. He was evaluated as PD again and got discharged. One month later, he was administrated with 2 cycles of CDK4/6 inhibitor (Palbociclib, 125 mg day 1–21) in combination with pembrolizumab (Keytruda, 200 mg day 1) but got limited response. Considering the patient’s strong desires and restricted options for further-line therapies, we treated him actively with 5 cycles of anlotinib (12 mg day 1–14), trastuzumab (Herceptin, 440 mg day 1) and pembrolizumab (Keytruda, 200 mg day 1). He responded rapidly and was assessed as a radiologic partial response (PR) after 4 cycles of treatment (Figure 2). He developed grade 3 oral mucositis requiring anlotinib reduced to 12 mg 2 doses every 3 days, and the symptom relieved within days. He presented durable responses and well tolerance to two further cycles of treatment. Until present, the patient only suffers grade I numbness of bilateral lower limbs without treatment interruption. He is alive with acceptable living quality for over 2 years since the initial diagnosis, and thus far the combination immunotherapy is ongoing. Complete treatment timeline is presented in Figure 3. Tumor markers are also monitored during the whole process of therapy (Figure 4).

Figure 2 Imaging findings of response after combination immunotherapy. (A) The abdominal CT scan showed a thickening wall of lesser curvature and multiple enlarged peri-gastric lymph nodes (54 mm × 47 mm). (B) Enlarged lymph nodes (28 mm × 28 mm) surrounding the celiac trunk. (C) Isolated intrahepatic nodule (18 mm × 14 mm). (D) Nodules in the lower lobe of the left lung (14 mm × 13 mm). (E) The thickening wall of lesser curvature and peri-gastric lymph nodes shrunk (26 mm × 22 mm). (F) Reduced size of lymph nodes (18 mm × 18 mm) surrounding the celiac trunk. (G) Reduced size of the intrahepatic nodule (7 mm × 5 mm). (H) Nodules in the lower lobe of the left lung grew down to almost disappeared (14 mm × 13 mm). (A) to (D) were generated before combination therapy (2019-4-10); (E) to (H) were generated after 2 cycles of combination immunotherapy (2019-5-28).
Figure 3 Timeline of the patient’s multiline treatments, treatment-related adverse effects and responses. PD, progressive disease; PR, partial response; RT, radiotherapy; GI, gastrointestinal; XELOX, oxaliplatin + capecitabine.
Figure 4 Dynamic change curves of tumor markers in peripheral blood after initiation of multiline treatment. (A) The CEA showed a fluctuation in the related quantity during the previous lines of treatment and peaking when initial ICI regimen failed, followed by a rapid decrease after ICI rechallenge. (B) and (C) The curves of NSE and CA-125 were similar to (A). The horizontal dashed line represented the normal range of tumor markers. Intuitively, tumor markers decreased to a low level and kept within a normal range, which indicated a favorable response. CEA, curves of carcinoembryonic antigen; NSE, neuron-specific enolase; CA-125, carbohydrate antigen-125.

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee(s) and with the Helsinki Declaration (as revised in 2013). Written informed consent was obtained from the patient for the publication of any potentially identifiable images or data included in this article. A copy of the written consent is available for review by the editorial office of this journal.


Discussion

In this report, we described a patient with late-stage GC who progressed on multiline therapies including ICI but showed a durable response to the combination of ICI rechallenge, RTKI and anti-HER2 therapy.

Locally advanced/metastatic (stage IV) GC is refractory and has challenged oncologists for years. First-line and second-line therapies have been standardized, resulting in relatively uniform regimens (2,4). In terms of this patient, the sequential regimen of oxaliplatin, capecitabine, irinotecan, taxanes and S-1 was adopted. Unfortunately, chemoradiotherapy was inefficacious and elicited severe treatment-related adverse effects (trAEs). We had no choices for evidence-based chemotherapies, therefore anchored hopes on target therapies.

Palbociclib is a highly selective CDK 4/6 inhibitor which arrests cell cycle progression at the G1 phase, thereby plays an anti-tumor role (10). According to the TCGA database, a portion of GC harbor abnormal cell-cycle-related molecules (11). Besides, in vitro studies also indicated that certain GC cells are sensitive to cell cycle inhibiting (12,13). These encouraging results drive us to experimentally commence the patient with Palbociclib.

In our case, the combination of trastuzumab, pembrolizumab and anlotinib eventually led to patient’s durable responses, based on serological and imaging evidence. It only brought about mild neurological adverse effects to the patient. Notably, this patient was initially insensitive to both immunotherapy and anti-HER2 therapy. More interestingly, anti-angiogenic single therapy only brings about poor response rate among advanced GC patients, and so anlotinib is typically used with other agents. Apparently, the unexpected durable response could not be elicited by either of these three drugs alone. The addition of anlotinib to ICIs is very likely to rejuvenate immunotherapy, which contributes to the success of the rechallenge.

The primary lesion of this patient was tested HER2 negative by FISH, thus was not generally indicated for anti-HER2 treatment due to the result of the ToGA trial (14,15). However, several factors were taken into considerations. First, the patient had previously completed a next-generation sequencing of the liver metastatic biopsy which suggested HER2 amplification. Literatures have reported the discordance of HER2 status between primary and metastatic lesions, which might be attributed to tumor heterogeneity (16,17). Second, the HER2 expression level in a single location could vary along with disease evolution or therapeutic process (17,18). Third, with the advent of new detection strategy, a portion of patients who are initially evaluated HER2 negative still benefit from anti-HER2 therapy (17,19). Therefore, we added trastuzumab into the combination regimen.

The combined administration of anti-PD-1 and anti-HER2 therapy has theoretical and practical foundations. Reportedly, HER2 blockade enhances antibody-dependent cell-mediated cytotoxicity (ADCC) via T cell priming, which has a synergistic effect with anti-PD-1 therapy (20). The 1188 PD study was a nonrandomized, phase II, multicohort trial to evaluate the efficacy and safety of pembrolizumab combined with margetuximab (a more efficient monoclonal antibody targeting HER2) in the treatment of HER2 positive GC after progression on first-line trastuzumab. The combination immunotherapy in this study resulted in a median OS of 12.9 months, longer than previous second-line data (9.6 months in Rainbow and 9.1 months in Keynote-061) (21). Notably, a considerable portion of patients who converted to HER2 negative after receiving trastuzumab still benefited from combination immunotherapy. These results suggest that concurrent anti-PD-1 and anti-HER2 therapy is a robust and efficacious combination strategy.

Anlotinib is a novel small-molecule RTKI which blockades VEGFR-2, PDGFR-β, FGFR-1 and c-Kit, posing a powerful anti-angiogenesis activity (22). Its efficacy and safety in the salvage treatment of solid tumors have been demonstrated by several clinical trials (22,23). The result of phase III ALTER 0303 trial showed that anlotinib monotherapy brought significant benefits in OS (9.6 vs. 6.3 months) and PFS (5.4 vs. 1.4 months) to patients with advanced non-small cell lung cancer over placebo (24). Recently in a phase I pioneer study, a combination of sintilimab (targeting PD-1) and anlotinib presented favorable efficacy and safety in the first-line therapy of lung cancer (25). In the field of GC, preclinical studies focusing on targeting both VEGFR-2 and PD-1 have also shown promising outcomes (26,27). Considering the current evidence and the promising outcome in our case, we have good reason to assume the extraordinary anti-tumor effect of this combination among GC patients.

Actually, this synergistic effect has been partly elucidated by previous studies. Angiogenesis, featured with aberrant blood vessels formation, is highly related to the progression of cancer and immunoregulation (28). Typically, VEGF signaling mediates the immunosuppressive tumor microenvironment (TME) by reducing functional T cell infiltration, inducing and aggregating suppressive immune cells within tumor (29). The administration of anti-angiogenic agents contributes to the normalization and maturation of aberrant blood vessels, which improves vascular permeability and therefore enhances the drug delivery, perfusion and oxygenation of TME (30). As the ameliorative TME facilitates T cell infiltration, ICI further mediates T cell mobilization and activation. In that way, host anti-tumor function could be restored.

Besides the aforementioned combinations (ICI + anti-HER2 or ICI + anti-angiogenesis), other combination strategies have also been explored presently. In the phase II study CheckMate-032, dual pathway ICIs (nivolumab targeting PD-1 and ipilimumab targeting CTLA-4) resulted in an ORR of 24% among advanced/metastatic gastroesophageal cancer patients who had previously treated with ≥1 chemotherapy regimen (31). In the Keynote-062 study, among MSI-H subgroup, patients who received concurrent immunotherapy and chemotherapy indicated significantly higher OS compared with chemotherapy alone (9). So far, evidence of various combination strategies is still insufficient. The optimal combination regimens, potential beneficiaries and the underlying molecular biology theories still need to be addressed in the future.

In the clinical setting, suspension and restart of ICI are also issues worthy of discussing. Treatment resistance and severe trAEs secondary to immunotherapy commonly lead to drug withdrawal. An encouraging fact is that ICI (alone or in combination with other therapies) rechallenge is indeed efficacious in certain situations (32-36). However, little information is known in the field of GC. It might be easier to comprehend the efficacy of ICI resume following adverse-event-related drug withdrawal, while acquired responses in the context of initial treatment failure remain incomprehensible. None of ICI, anlotinib or trastuzumab alone could lead to a favorable response. We speculate that the reversal of immunosuppressive TME mediated by anlotinib and the enhanced ADCC effect mediated by trastuzumab successfully broke the tumor resistance to ICI and therefore contributed to the favorable anti-tumor response.

This case indicates that previously ICI-treated GC patients might regain sensitivity to ICI. It has significant reference value for the clinical management of GC patients. Future investigations should focus on the rationales and applications of combination immunotherapy and ICI rechallenge.


Acknowledgments

Funding: This work was supported by the National Key Research and Development Program of China [2017YFC1308900, 2017YFC0908400]; National Natural Science Foundation of China [81602057]; Clinical Medicine Plus X - Young Scholars Project of Peking University [PKU2019LCXQ020, PKU2018LCXQ018]; and Beijing Municipal Administration of Hospital’s Youth Program [20171102].


Footnote

Reporting Checklist: The authors have completed the CARE reporting checklist. Available at https://apm.amegroups.com/article/view/10.21037/apm-21-83/rc

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://apm.amegroups.com/article/view/10.21037/apm-21-83/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee(s) and with the Helsinki Declaration (as revised in 2013). Written informed consent was obtained from the patient for the publication of any potentially identifiable images or data included in this article. A copy of the written consent is available for review by the editorial office of this journal.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


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;68:394-424. [Crossref] [PubMed]
  2. Smyth EC, Moehler M. Late-line treatment in metastatic gastric cancer: today and tomorrow. Ther Adv Med Oncol 2019;11:1758835919867522. [Crossref] [PubMed]
  3. Chan WL, Lam KO, So TH, et al. Third-line systemic treatment in advanced/metastatic gastric cancer: a comprehensive review. Ther Adv Med Oncol 2019;11:1758835919859990. [Crossref] [PubMed]
  4. Smyth EC, Verheij M, Allum W, et al. Gastric cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 2016;27:v38-49. [Crossref] [PubMed]
  5. Okazaki T, Honjo T. PD-1 and PD-1 ligands: from discovery to clinical application. Int Immunol 2007;19:813-24. [Crossref] [PubMed]
  6. 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;25:6160-9. [Crossref] [PubMed]
  7. Mishima S, Kawazoe A, Nakamura Y, et al. Clinicopathological and molecular features of responders to nivolumab for patients with advanced gastric cancer. J Immunother Cancer 2019;7:24. [Crossref] [PubMed]
  8. Shitara K, Özgüroğlu M, Bang Y, et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet 2018;392:123-33. [Crossref] [PubMed]
  9. Tabernero J, Van Cutsem E, Bang Y, et al. Pembrolizumab with or without chemotherapy versus chemotherapy for advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma: the phase III KEYNOTE-062 study. J Clin Oncol 2019;37:A4007. [Crossref]
  10. Goel S, DeCristo MJ, Watt AC, et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature 2017;548:471-5. [Crossref] [PubMed]
  11. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014;513:202-9. [Crossref] [PubMed]
  12. Valenzuela CA, Vargas L, Martinez V, et al. Palbociclib-induced autophagy and senescence in gastric cancer cells. Exp Cell Res 2017;360:390-6. [Crossref] [PubMed]
  13. Min A, Kim JE, Kim Y, et al. Cyclin E overexpression confers resistance to the CDK4/6 specific inhibitor palbociclib in gastric cancer cells. Cancer Lett 2018;430:123-32. [Crossref] [PubMed]
  14. Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 2010;376:687-97. Erratum in: Lancet 2010;376:1302. [Crossref] [PubMed]
  15. Hofmann M, Stoss O, Shi D, et al. Assessment of a HER2 scoring system for gastric cancer: results from a validation study. Histopathology 2008;52:797-805. [Crossref] [PubMed]
  16. Gumusay O, Benekli M, Ekinci O, et al. Discordances in HER2 status between primary gastric cancer and corresponding metastatic sites. Jpn J Clin Oncol 2015;45:416-21. [Crossref] [PubMed]
  17. Park SR, Park YS, Ryu M, et al. Extra-gain of HER2-positive cases through HER2 reassessment in primary and metastatic sites in advanced gastric cancer with initially HER2-negative primary tumours: results of GASTric cancer HER2 reassessment study 1 (GASTHER1). Eur J Cancer 2016;53:42-50. [Crossref] [PubMed]
  18. Saeki H, Oki E, Kashiwada T, et al. Re-evaluation of HER2 status in patients with HER2-positive advanced or recurrent gastric cancer refractory to trastuzumab (KSCC1604). Eur J Cancer 2018;105:41-9. [Crossref] [PubMed]
  19. Wang H, Li B, Liu Z, et al. HER2 copy number of circulating tumour DNA functions as a biomarker to predict and monitor trastuzumab efficacy in advanced gastric cancer. Eur J Cancer 2018;88:92-100. [Crossref] [PubMed]
  20. Stagg J, Loi S, Divisekera U, et al. Anti-ErbB-2 mAb therapy requires type I and II interferons and synergizes with anti-PD-1 or anti-CD137 mAb therapy. Proc Natl Acad Sci USA 2011;108:7142-7. [Crossref] [PubMed]
  21. Catenacci DV, Park H, Uronis H, et al. 1188PD Margetuximab (M) + pembrolizumab (P) for treatment of patients (pts) with HER2+ gastroesophageal adenocarcinoma (GEA) post-trastuzumab (T): survival analysis. Ann Oncol 2019;30:z214-53. [Crossref]
  22. Shen G, Zheng F, Ren D, et al. Anlotinib:a novel multi-targeting tyrosine kinase inhibitor in clinical development. J Hematol Oncol 2018;11:120. [Crossref] [PubMed]
  23. Syed YY. Anlotinib: First Global Approval. Drugs 2018;78:1057-62. [Crossref] [PubMed]
  24. Han B, Li K, Wang Q, et al. Effect of anlotinib as a third-line or further treatment on overall survival of patients with advanced non–small l cell lung cancer. JAMA Oncol 2018;4:1569. [Crossref] [PubMed]
  25. Han B, Chu T, Zhong R, et al. P1.04-02 Efficacy and safety of sintilimab with anlotinib as first-line therapy for advanced non-small cell lung cancer (NSCLC). J Thorac Oncol 2019;14:S439. [Crossref]
  26. Hironaka S. Anti-angiogenic therapies for gastric cancer. Asia Pac J Clin Oncol 2019;15:208-17. [PubMed]
  27. Chen LT, Oh DY, Ryu MH, et al. Anti-angiogenic therapy in patients with advanced gastric and gastroesophageal junction cancer: a systematic review. Cancer Res Treat 2017;49:851-68. [Crossref] [PubMed]
  28. Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature 2000;407:249-57. [Crossref] [PubMed]
  29. Manegold C, Dingemans AC, Gray JE, et al. The potential of combined immunotherapy and antiangiogenesis for the synergistic treatment of advanced NSCLC. J Thorac Oncol 2017;12:194-207. [Crossref] [PubMed]
  30. Huang Y, Yuan J, Righi E, et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc Natl Acad Sci U S A 2012;109:17561-6. [Crossref] [PubMed]
  31. Janjigian YY, Bendell J, Calvo E, et al. CheckMate-032 study: efficacy and safety of nivolumab and nivolumab plus ipilimumab in patients with metastatic esophagogastric cancer. J Clin Oncol 2018;36:2836-44. Erratum in: J Clin Oncol 2019;37:443. [Crossref] [PubMed]
  32. Tedbirt B, De Pontville M, Branger P, et al. Rechallenge of immune checkpoint inhibitor after pembrolizumab-induced myasthenia gravis. Eur J Cancer 2019;113:72-4. [Crossref] [PubMed]
  33. Watanabe H, Kubo T, Ninomiya K, et al. The effect and safety of immune checkpoint inhibitor rechallenge in non-small cell lung cancer. Jpn J Clin Oncol 2019;49:762-5. [Crossref] [PubMed]
  34. Winer A, Ghatalia P, Bubes N, et al. Dual checkpoint inhibition with ipilimumab plus nivolumab after progression on sequential PD-1/PDL-1 inhibitors pembrolizumab and atezolizumab in a patient with Lynch Syndrome, metastatic colon, and localized urothelial cancer. Oncologist 2019;24:1416-9. [Crossref] [PubMed]
  35. Kato J, Hida T, Kamiya T, et al. Rechallenge with nivolumab after vemurafenib treatment of initially nivolumab-resistant advanced melanoma. JAMA Dermatol 2018;154:621-2. [Crossref] [PubMed]
  36. Spain L, Walls G, Messiou C, et al. Efficacy and toxicity of rechallenge with combination immune checkpoint blockade in metastatic melanoma: a case series. Cancer Immunol Immunother 2017;66:113-7. [Crossref] [PubMed]
Cite this article as: Zhang Z, Cheng S, Qi C, Zhang X, Peng Z, Shen L. Response to the rechallenge of combination immunotherapy in a patient with late-stage gastric cancer: case report. Ann Palliat Med 2022;11(2):818-826. doi: 10.21037/apm-21-83

Download Citation