Neoadjuvant and Adjuvant Therapy for Gastric Cancer: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , ,

Gastric cancer is a deadly disease with worldwide prevalence that is often diagnosed at late stages. About two-thirds of patients in Western countries will present with locally advanced gastric cancer (LAGC). When patients are diagnosed with LAGC, they frequently undergo surgery and perioperative chemotherapy. However, the most effective multimodality treatment regimen for LAGC has yet to be determined. In aiming to improve outcomes, current trials are examining immunotherapies and targeted therapies based on a growing understanding of the unique molecular characteristics and subtypes of LAGC.

  • gastric cancer
  • neoadjuvant therapy
  • adjuvant therapy

1. Introduction

Gastric cancer (GC) represents the world’s fifth most common cancer and fourth leading cause of cancer death, with 1.1 million new cases and 770,000 deaths estimated in 2020 [1][2]. The incidence of GC is higher in males than in females in both developed and developing countries [1]. Incidence rates are highest in Eastern and Central Asia and Latin America; the average incidence rate of GC in Eastern Asia is 32.1 per 100,000 in males and 13.2 per 100,000 in females [3]. The incidence of GC peaks in the seventh and eighth decades of life [4]. Risk factors for non-cardia GC are Helicobacter pylori (H. pylori) infection, high-salt diets, smoking, prior gastric surgery, and family history [4]. Cardia GC is associated with gastroesophageal reflux disease (GERD) and obesity [5]. The incidence of non-cardia GC has decreased worldwide over the past 50 years; however, cardia GC has increased sevenfold, especially in developed countries, and an unexpected increase in the incidence of GC has been seen in younger age groups (<50 years old) [6][7][8]. The overall decrease in non-cardia GC may be due to improved H. pylori treatments and advancements in food hygiene in recent decades [9].
The most common presenting symptoms of GC are non-specific weight loss, dyspepsia, abdominal pain, anorexia, and dysphagia [10]. Given the insidious nature of these symptoms, a majority of GC patients present with advanced or metastatic disease [11]. Therefore, mortality rates are high in GC, with patients presenting with unresectable or metastatic GC having a median survival of about one year with medical treatment [12]. Approximately two-thirds of patients in Western countries will present with locally advanced gastric cancer (LAGC), which is usually defined as the primary tumor extending beyond the muscularis propria (cT3-T4) or having nodal metastases (cN+) disease and without distant metastases (cM0) [11][13]. In contrast, the majority of patients in Japan and Republic of Korea present with early disease due to nationwide screening programs [14][15].

2. Molecular Subtypes and Biomarkers of GC

GC is a heterogenous disease. Over the past decade, an in-depth understanding of the molecular characteristics of GC has led to the advent of novel diagnostic and prognostic biomarkers as well as the development of targeted therapies. With precision medicine and an increasing ability to predict responders to various therapies, neoadjuvant and adjuvant therapies can be better utilized in GC patients.
In 2014, The Cancer Genome Atlas (TCGA) outlined a molecular classification system separating GC into four subtypes: tumors with high microsatellite instability (MSI-H), tumors that are Epstein–Barr virus positive (EBV+), genomically stable (GS) tumors, and tumors with chromosomal instability (CIN) [16]. These subtypes each have distinct genomic features, which may serve as targets for therapeutic agents. In 2015, the Asian Cancer Research Group published their own molecular classification of GC with associated prognostic differences: tumors with microsatellite instability (MSI), mesenchymal tumors (microsatellite-stable tumors with epithelial-to-mesenchymal transition phenotype (MSS/EMT)), and non-EMT MSS tumors that are p53+ and p53− [17]. Patients with MSS/EMT-type GC were younger and had the worst prognosis compared to patients with the other subtypes. In contrast, patients with MSI-H tumors had the best prognosis; the MSS/p53+ genotype was closely linked to EBV+ status, and patients with MSS/p53− tumors had a better prognosis than those with MSS/p53+ tumors.
MSI-H tumors have a high tumor mutational burden and are deficient in one or more mismatch repair proteins, i.e., mismatch repair deficient (dMMR). dMMR is a surrogate for the MSI-H phenotype. Polymerase chain reaction can be used to detect amplified microsatellite loci and is more accurate than IHC, though more expensive [18]. MSI-H tumors are associated with older age and tend to occur in the distal stomach [19]. About 10–20% of GCs are MSI-H [20][21]. MSI-H tumors tend to have fewer lymph node metastases and better OS [22][23]. In addition, MSI-H GCs frequently express immune checkpoint molecules like PD-L1, making them possibly eligible for treatment with immune checkpoint inhibitors (ICIs) [24]. MSI-H GCs can be identified using immunohistochemistry (IHC) with antibodies directed at mismatch repair proteins: MLH1, PMS2, MSH2, and MSH6 [23].
The EBV+ subtype of GC accounts for 10% of cases and frequently harbors PIK3CA and ARID1A mutations as well as amplification of JAK2, PD-L1, and PD-L2 and DNA hypermethylation [16]. Testing for EBV infection is best implemented by EBV encoding region (EBER) in situ hybridization, which would show a nuclear signal [25]. EBV-associated GC is more common in younger men and occurs primarily in the proximal stomach or postsurgical gastric stump [26]. The EBV+ subtype of GC generally has a more favorable prognosis and fewer lymph node metastases than non-EBV GC [27]. In addition, given that the EBV+ subtype of GC often is associated with modulation of the tumor microenvironment (TME) to evade the host immune response (such as by upregulation of PD-L1 expression by CD274 focal amplification and IFN-γ-mediated signaling, causing immunosuppression), immune checkpoint blockade arose as a promising avenue of treatment [28][29][30].
HER2 gene amplification occurs in 10–30% of GCs, which results in overexpression of HER2, a receptor tyrosine kinase [16]. HER2 status is mainly assessed through IHC and/or fluorescence in situ hybridization (FISH). Given its relatively low cost and ease of performance, IHC is typically used first to test for HER2 overexpression. A 3+ result is considered positive. If the results are equivocal (2+), FISH is performed [31]. HER2+ status is prognostic of poorer outcomes in GC, so identification is critical and may predict which patients will benefit from the addition of trastuzumab or other HER2-directed therapies to their chemotherapy regimen [32][33].
Finally, PD-L1 IHC is routinely performed to help predict potential benefit from immunotherapy. In addition, the EBV+ and MSI-H subtypes are associated with the overexpression of PD-LI in tumor, stromal, and immune cells [16]. The combined positive score (CPS) quantifies PD-LI expression in tumor and tumor-associated immune cells, where CPS ≥ 1 is considered positive [34]. A higher score correlates with an increased probability of clinical benefit from PD-L1 inhibition [35]. An increasing number of studies have been conducted to investigate the efficacy of immunotherapy for LAGC patients, which will be discussed later.

3. Immune Checkpoint Blockade for MSI-H LAGC

As mentioned above, MSI-H LAGC is associated with an improved prognosis [16][36][37]. In the MAGIC trial, which established perioperative chemotherapy as the standard of care for non-Asian patients with LAGC, patients with MSI-H tumors had an improved prognosis compared to patients with microsatellite-stable (MSS) tumors in the surgery-only group (hazard ratio (HR) 0.35, 95% confidence interval (CI) 0.11–1.11, p = 0.08). Perioperative chemotherapy and surgery in the MSI-H subset of patients was associated with a worse OS compared to surgery alone (HR = 2.22, 95% CI 1.02–4.85, p = 0.04) [36][38]. In the CLASSIC trial, which found that adjuvant capecitabine and oxaliplatin after curative D2 gastrectomy is effective in Asian LAGC patients, patients with MSI-H tumors did not have a survival benefit when adjuvant chemotherapy was added to surgery (5-year disease-free survival 83.5% versus 85.7%, p = 0.931) [37][39]. A pooled meta-analysis of four neoadjuvant/adjuvant chemotherapy trials (MAGIC, CLASSIC, ARTIST, ITACA-S) found that only the non-MSI-H LAGC patients benefited from chemotherapy and surgery compared to surgery only, with a five-year OS of 62% versus 53% (HR = 0.75, 95% CI 0.60–0.94) [40]. Thus, neoadjuvant/adjuvant chemotherapy is likely ineffective for MSI-H LAGC.
However, there may be a role for neoadjuvant or adjuvant immunotherapy, especially in patients with MSI-H tumors. The TME plays a key role in GC progression through immune metabolic reprogramming and alteration of immune cells to evade host defenses [41]. The immune TME in GC contains T and B lymphocytes, natural killer (NK) cells, macrophages, and neutrophils [42]. These cells can promote or inhibit tumor immunity. GC exerts a variety of mechanisms to evade the host immune system. For instance, tumors can release anti-inflammatory cytokines such as IL-10 and TGF-β, which can cause the recruitment of regulatory T cells (Tregs) and inhibit the antitumor effect of lymphocytes [42][43]. Tregs can cause immune suppression through multiple mechanisms, including CTLA-4 expression to inhibit antigen-presenting cells [44]. In addition, GCs themselves can express PD-L1, which interacts with PD-1 on the cell surface of T cells, causing anergy and/or apoptosis of T cells [45]. With this knowledge, ICIs have become a promising area of therapy for many tumor types including GC.
Several studies show an improved response to ICIs when combined with chemotherapy in a variety of cancers, such as non-small-cell lung cancer, mesothelioma, and renal cell carcinoma [46][47][48]. This synergistic effect is thought to be due to chemotherapy activating an endogenous antitumor immune response, causing the enhancement of co-stimulatory molecules like CD80 and CD86 and the downregulation of PD-L1, and inducing immunogenic tumor cell death [49]. The current Food and Drug Administration (FDA)-approved ICIs target two key T-cell signaling pathways: PD-L1/PD-1 and CTLA-4. Although PD-L1 expression in GC has been associated with poorer outcomes, anti-PD-L1 antibodies (e.g., atezolizumab, avelumab, durvalumab) and anti-PD-1 antibodies (e.g., nivolumab, pembrolizumab) have been shown to improve OS in patients with advanced or metastatic GC [50][51]. CTLA-4 inhibitors such as tremelimumab and ipilimumab have also been explored as options for the treatment of GC. The KEYNOTE-062 phase III trial randomized patients to pembrolizumab alone, pembrolizumab and chemotherapy (5-FU and oxaliplatin), and chemotherapy alone in patients with advanced GC and PD-L1 CPS ≥ 1. That study found that pembrolizumab was as effective as chemotherapy with fewer treatment-related adverse effects [52]. Overall, pembrolizumab plus chemotherapy was not more effective than chemotherapy alone [52]. Post hoc analysis of the KEYNOTE-062 trial, along with the KEYNOTE-059 and KEYNOTE-061 trials, found a significant benefit for pembrolizumab in the subset of patients with MSI-H tumors [53]. One meta-analysis analyzed the RCTs that studied first-line advanced GC treatment with chemotherapy plus ICIs vs. chemotherapy alone [54]. That analysis found a significant reduction in the risk of death and progression in those treated with combination chemotherapy plus ICIs, across patients with PD-L1 CPS ≥ 10 and CPS ≥ 1, but a larger reduction in risk of death and progression was seen in patients with CPS ≥ 10.
Given the efficacy of PD-1/PD-L1 inhibition in MSI-H advanced GC, the question arose of whether similar effects would be seen in MSI-H LAGC. The GERCOR NEONIPIGA phase II trial evaluated the pathological complete response (pCR) rate after perioperative chemotherapy in GC/gastroesophageal junction adenocarcinoma (GEJC) with dMMR/MSI-H status [55]. Specifically, 32 patients with cT2-T4/NX/M0 resectable dMMR/MSI-H GC/GEJC underwent neoadjuvant nivolumab and ipilimumab for four cycles followed by surgery and adjuvant nivolumab for nine cycles [55]. Overall, 59% of tumors had complete regression with no residual tumor (TRG1a) and 14% had less than 10% residual tumor (TRG1b) [55][56]. At the most recent follow up of those patients who achieved Becker TRG 1a/1b, no patients had died or had recurrence. The authors concluded that neoadjuvant and adjuvant nivolumab and ipilimumab was safe and feasible in patients with MSI-H LAGC [55].
The phase II INFINITY trial enrolled patients with MSI-H cT2 or greater (any N, M0) GC/GEJC (n = 15) and treated these patients with tremelimumab (anti-CTLA-4) and durvalumab (anti-PD-L1) for one cycle followed by durvalumab every four weeks for two cycles followed by surgery [57]. Ultimately, 60% of patients achieved pCR, and 80% had <10% viable tumor. Notably, only one out of seven patients with T4 tumors achieved pCR [57]. In summary, neoadjuvant and adjuvant immunotherapy with anti-PD-L1 and anti-CTLA-4 agents has demonstrated promising tumor-eradicating activity in patients with dMMR/MSI-H LAGC.

4. Immune Checkpoint Blockade Plus Chemotherapy for Non-MSI-H LAGC

Although immune checkpoint blockade has been shown to be effective in MSI-H LAGC, most LAGCs are MSS [21]. ICIs in combination with chemotherapy may be effective in non-MSI-H LAGC based on studies in advanced patients. The CheckMate-649 phase III trial found that, patients with advanced GC/GEJC treated with nivolumab, an anti-PD-1 antibody, obtained a survival benefit when combined with chemotherapy compared to chemotherapy alone in patients with tumors with PD-L1 CPS ≥ 1 and ≥5 [35]. This led to the FDA approval of nivolumab in April 2021 for use in advanced GC/GEJC regardless of PD-L1 CPS, though the National Comprehensive Cancer Network (NCCN) guidelines specify their category 1 designation for CPS ≥ 5 [58]. The CheckMate-577 trial showed that nivolumab can also prolong survival compared to placebo in the adjuvant setting in patients with resected stage II and III esophageal or GEJC who also received neoadjuvant chemoradiotherapy [59]. Recent data from key phase III RCTs performed in China, Rationale-305 and ORIENT-16, show an OS benefit in patients with PD-L1+ unresectable GC/GEJC treated with PD-L1 inhibitors in combination with chemotherapy compared to placebo and chemotherapy [60][61]. Rationale-305 found that the PD-L1+ group (defined as CPS ≥ 5) treated with tislelizumab and chemotherapy had improved progression-free survival (PFS) and median OS (17.2 vs. 12.6 months, one-sided p-value = 0.0056) [61]. ORIENT-16 demonstrated superior OS with sintilimab and chemotherapy in all patients regardless of PD-L1 status (31.9% risk reduction, p < 0.0001), with a stronger reduction in patients with CPS ≥ 5 (41.3% risk reduction, p < 0.0001) [60]. ATTRACTION-4, a phase II-III multicenter trial across Japan, Republic of Korea, and Taiwan, randomized patients with unresectable advanced or recurrent GC/GEJC to either nivolumab with chemotherapy or placebo and chemotherapy; they found that the addition of nivolumab significantly improved PFS (10.45 vs. 8.34 months, p = 0.0007) but not OS [62].
Several studies are underway to investigate the efficacy of ICIs with neoadjuvant and/or adjuvant chemotherapy for non-MSI-H LAGC. A single-arm phase II trial (NCT0291816) recruited 34 patients with LAGC/GEJC; these patients were treated with capecitabine and oxaliplatin (CAPOX) before and after surgery as well as one cycle of pembrolizumab immediately before surgery followed by twelve months of maintenance pembrolizumab [63]. It was found that 20.6% of patients achieved pCR, surpassing the target pCR of 15% [63]. DANTE is a multicenter phase II trial that is comparing perioperative atezolizumab (anti-PD-L1) and FLOT chemotherapy against FLOT alone in patients with operable LAGC regardless of PD-L1 status (≥cT2 or N+ GC/GEJC) [64]. The preliminary results show an increase in pathological regression rates in patients who received both atezolizumab and FLOT, particularly in patients with higher PD-L1 expression (overall pT0 23% vs. 15%, pN0 68% vs. 54%). The estimated accrual completion date is 2025. In addition, the KEYNOTE-585 trial is underway to investigate the efficacy and safety of neoadjuvant and adjuvant pembrolizumab and chemotherapy (either FLOT, cisplatin and 5-FU, or cisplatin and capecitabine) compared to placebo and chemotherapy in patients with stage II-IVa GC or GEJC [65]. The primary endpoints are OS, event-free survival, and pCR rate [65]. KEYNOTE-585 is estimated to complete accrual in June 2024 and will hopefully further elucidate the role of adjuvant immunotherapy in non-MSI-H LAGC. Finally, the MATTERHORN trial is currently recruiting around 900 patients with resectable stage II or higher GC/GEJC. These patients will be randomly assigned to receive either neoadjuvant durvalumab or placebo in combination with FLOT chemotherapy, followed by either adjuvant durvalumab or placebo monotherapy after surgical resection [66]. Durvalumab in combination with FLOT has been shown to have the potential to improve outcomes in two smaller clinical studies in patients with advanced GC/GEJC, hence leading to the development of MATTERHORN [67][68]. Expected accrual completion is in 2025.
Recent studies suggest that the timing of immunotherapy with chemotherapy may be critical the in treatment of GC. ATTRACTION-5 is a phase III trial of Asian patients with stage III GC/GEJC randomized to receive either nivolumab with adjuvant chemotherapy or placebo with adjuvant chemotherapy [69]. There was no improvement in OS, and the primary endpoint of PFS was not met (HR = 0.90, p = 0.4363) [69]. A phase II randomized trial (NCT04250948) evaluated the efficacy of the addition of toripalimab to perioperative chemotherapy in patients with LAGC/GEJC [70]. In the group receiving PD-L1 inhibitors, toripalimab was added preoperatively and as a maintenance adjuvant therapy. Patients in the toripalimab plus chemotherapy arm achieved a higher proportion of pCR (24.1% vs. 9.3%, p = 0.039) and TRG 0/1 (44.4% vs. 20.4%, p = 0.009) than patients in the chemotherapy arm [70]. Although these studies were conducted in different populations, together they suggest that perioperative immunotherapy may be more effective than adjuvant only when combined with chemotherapy. The upcoming results of MATTERHORN and KEYNOTE-585 will further inform the understanding of the role of immunotherapy in LAGC.

This entry is adapted from the peer-reviewed paper 10.3390/cancers15164114

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249.
  2. Morgan, E.; Arnold, M.; Camargo, M.C.; Gini, A.; Kunzmann, A.T.; Matsuda, T.; Meheus, F.; Verhoeven, R.H.A.; Vignat, J.; Laversanne, M.; et al. The Current and Future Incidence and Mortality of Gastric Cancer in 185 Countries, 2020–2040: A Population-Based Modelling Study. EClinicalMedicine 2022, 47, 101404.
  3. Balakrishnan, M.; George, R.; Sharma, A.; Graham, D.Y. Changing Trends in Stomach Cancer throughout the World. Curr. Gastroenterol. Rep. 2017, 19, 36.
  4. Smyth, E.C.; Nilsson, M.; Grabsch, H.I.; van Grieken, N.C.; Lordick, F. Gastric Cancer. Lancet 2020, 396, 635–648.
  5. Karimi, P.; Islami, F.; Anandasabapathy, S.; Freedman, N.D.; Kamangar, F. Gastric Cancer: Descriptive Epidemiology, Risk Factors, Screening, and PreventionGastric Cancer. Cancer Epidemiol. Biomark. Prev. 2014, 23, 700–713.
  6. Arnold, M.; Park, J.Y.; Camargo, M.C.; Lunet, N.; Forman, D.; Soerjomataram, I. Is Gastric Cancer Becoming a Rare Disease? A Global Assessment of Predicted Incidence Trends to 2035. Gut 2020, 69, 823–829.
  7. Rawla, P.; Barsouk, A. Epidemiology of Gastric Cancer: Global Trends, Risk Factors and Prevention. Przegląd Gastroenterol. 2019, 14, 26–38.
  8. Thrift, A.P.; Wenker, T.N.; El-Serag, H.B. Global Burden of Gastric Cancer: Epidemiological Trends, Risk Factors, Screening and Prevention. Nat. Rev. Clin. Oncol. 2023, 20, 338–349.
  9. Fuccio, L.; Eusebi, L.H.; Bazzoli, F. Gastric Cancer, Helicobacter Pylori Infection and Other Risk Factors. World J. Gastrointest. Oncol. 2010, 2, 342–347.
  10. Wanebo, H.J.; Kennedy, B.J.; Chmiel, J.; Steele, G., Jr.; Winchester, D.; Osteen, R. Cancer of the Stomach. A Patient Care Study by the American College of Surgeons. Ann. Surg. 1993, 218, 583–592.
  11. Hundahl, S.A.; Phillips, J.L.; Menck, H.R. The National Cancer Data Base Report on Poor Survival of U.S. Gastric Carcinoma Patients Treated with Gastrectomy: Fifth Edition American Joint Committee on Cancer Staging, Proximal Disease, and the “Different Disease” Hypothesis. Cancer 2000, 88, 921–932.
  12. Wagner, A.D.; Syn, N.L.; Moehler, M.; Grothe, W.; Yong, W.P.; Tai, B.-C.; Ho, J.; Unverzagt, S. Chemotherapy for Advanced Gastric Cancer. Cochrane Database Syst. Rev. 2017, 8, CD004064.
  13. Couto, E.; Marques, A.; Freitas, D.; Nabico, R. Locally Advanced Gastric Cancer: Current and Future Strategies to Improve Outcomes with Multimodality Approach. Surg. Gastroenterol. Oncol. 2020, 25, 17.
  14. Nashimoto, A.; Akazawa, K.; Isobe, Y.; Miyashiro, I.; Katai, H.; Kodera, Y.; Tsujitani, S.; Seto, Y.; Furukawa, H.; Oda, I.; et al. Gastric Cancer Treated in 2002 in Japan: 2009 Annual Report of the JGCA Nationwide Registry. Gastric Cancer 2013, 16, 1–27.
  15. Jung, K.-W.; Won, Y.-J.; Kong, H.-J.; Oh, C.-M.; Shin, A.; Lee, J.-S. Survival of Korean Adult Cancer Patients by Stage at Diagnosis, 2006-2010: National Cancer Registry Study. Cancer Res. Treat. 2013, 45, 162–171.
  16. The Cancer Genome Atlas Research Network. Comprehensive Molecular Characterization of Gastric Adenocarcinoma. Nature 2014, 513, 202–209.
  17. Cristescu, R.; Lee, J.; Nebozhyn, M.; Kim, K.-M.; Ting, J.C.; Wong, S.S.; Liu, J.; Yue, Y.G.; Wang, J.; Yu, K.; et al. Molecular Analysis of Gastric Cancer Identifies Subtypes Associated with Distinct Clinical Outcomes. Nat. Med. 2015, 21, 449–456.
  18. Chen, M.-L.; Chen, J.-Y.; Hu, J.; Chen, Q.; Yu, L.-X.; Liu, B.-R.; Qian, X.-P.; Yang, M. Comparison of Microsatellite Status Detection Methods in Colorectal Carcinoma. Int. J. Clin. Exp. Pathol. 2018, 11, 1431–1438.
  19. Guan, W.-L.; Ma, Y.; Cui, Y.-H.; Liu, T.-S.; Zhang, Y.-Q.; Zhou, Z.-W.; Xu, J.-Y.; Yang, L.-Q.; Li, J.-Y.; Sun, Y.-T.; et al. The Impact of Mismatch Repair Status on Prognosis of Patients With Gastric Cancer: A Multicenter Analysis. Front. Oncol. 2021, 11, 712760.
  20. Beghelli, S.; de Manzoni, G.; Barbi, S.; Tomezzoli, A.; Roviello, F.; Di Gregorio, C.; Vindigni, C.; Bortesi, L.; Parisi, A.; Saragoni, L.; et al. Microsatellite Instability in Gastric Cancer Is Associated with Better Prognosis in Only Stage II Cancers. Surgery 2006, 139, 347–356.
  21. Fang, W.-L.; Chang, S.-C.; Lan, Y.-T.; Huang, K.-H.; Chen, J.-H.; Lo, S.-S.; Hsieh, M.-C.; Li, A.F.-Y.; Wu, C.-W.; Chiou, S.-H. Microsatellite Instability Is Associated with a Better Prognosis for Gastric Cancer Patients after Curative Surgery. World J. Surg. 2012, 36, 2131–2138.
  22. Cortes-Ciriano, I.; Lee, S.; Park, W.-Y.; Kim, T.-M.; Park, P.J. A Molecular Portrait of Microsatellite Instability across Multiple Cancers. Nat. Commun. 2017, 8, 15180.
  23. Mathiak, M.; Warneke, V.S.; Behrens, H.-M.; Haag, J.; Böger, C.; Krüger, S.; Röcken, C. Clinicopathologic Characteristics of Microsatellite Instable Gastric Carcinomas Revisited: Urgent Need for Standardization. Appl. Immunohistochem. Mol. Morphol. 2017, 25, 12–24.
  24. Zhao, P.; Li, L.; Jiang, X.; Li, Q. Mismatch Repair Deficiency/Microsatellite Instability-High as a Predictor for Anti-PD-1/PD-L1 Immunotherapy Efficacy. J. Hematol. Oncol. 2019, 12, 54.
  25. Chen, X.-Z.; Chen, H.; Castro, F.A.; Hu, J.-K.; Brenner, H. Epstein-Barr Virus Infection and Gastric Cancer: A Systematic Review. Medicine 2015, 94, e792.
  26. Murphy, G.; Pfeiffer, R.; Camargo, M.C.; Rabkin, C.S. Meta-Analysis Shows that Prevalence of Epstein-Barr Virus-Positive Gastric Cancer Differs Based on Sex and Anatomic Location. Gastroenterology 2009, 137, 824–833.
  27. Constanza Camargo, M.; Kim, W.-H.; Chiaravalli, A.M.; Kim, K.-M.; Corvalan, A.H.; Matsuo, K.; Yu, J.; Sung, J.J.Y.; Herrera-Goepfert, R.; Meneses-Gonzalez, F.; et al. Improved Survival of Gastric Cancer with Tumour Epstein–Barr Virus Positivity: An International Pooled Analysis. Gut 2014, 63, 236–243.
  28. Bai, Y.; Xie, T.; Wang, Z.; Tong, S.; Zhao, X.; Zhao, F.; Cai, J.; Wei, X.; Peng, Z.; Shen, L. Efficacy and Predictive Biomarkers of Immunotherapy in Epstein-Barr Virus-Associated Gastric Cancer. J. Immunother. Cancer 2022, 10, e004080.
  29. Bauer, M.; Jasinski-Bergner, S.; Mandelboim, O.; Wickenhauser, C.; Seliger, B. Epstein–Barr Virus—Associated Malignancies and Immune Escape: The Role of the Tumor Microenvironment and Tumor Cell Evasion Strategies. Cancers 2021, 13, 5189.
  30. Nakano, H.; Saito, M.; Nakajima, S.; Saito, K.; Nakayama, Y.; Kase, K.; Yamada, L.; Kanke, Y.; Hanayama, H.; Onozawa, H.; et al. PD-L1 Overexpression in EBV-Positive Gastric Cancer is Caused by Unique Genomic or Epigenomic Mechanisms. Sci. Rep. 2021, 11, 1982.
  31. Hoang, M.P.; Sahin, A.A.; Ordòñez, N.G.; Sneige, N. HER-2/Neu Gene Amplification Compared with HER-2/Neu Protein Overexpression and Interobserver Reproducibility in Invasive Breast Carcinoma. Am. J. Clin. Pathol. 2000, 113, 852–859.
  32. Trumbull, D.A.; Lemini, R.; Díaz Vico, T.; Jorgensen, M.S.; Attwood, K.; Ji, W.; Brady, M.; Gabriel, E.; Kukar, M. Prognostic Significance of Complete Pathologic Response Obtained with Chemotherapy Versus Chemoradiotherapy in Gastric Cancer. Ann. Surg. Oncol. 2021, 28, 766–773.
  33. Bang, Y.-J.; Van Cutsem, E.; Feyereislova, A.; Chung, H.C.; Shen, L.; Sawaki, A.; Lordick, F.; Ohtsu, A.; Omuro, Y.; Satoh, T.; 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, 687–697.
  34. Ghidini, M.; Petrillo, A.; Botticelli, A.; Trapani, D.; Parisi, A.; La Salvia, A.; Sajjadi, E.; Piciotti, R.; Fusco, N.; Khakoo, S. How to Best Exploit Immunotherapeutics in Advanced Gastric Cancer: Between Biomarkers and Novel Cell-Based Approaches. J. Clin. Med. Res. 2021, 10, 1412.
  35. Janjigian, Y.Y.; Shitara, K.; Moehler, M.; Garrido, M.; Salman, P.; Shen, L.; Wyrwicz, L.; Yamaguchi, K.; Skoczylas, T.; Campos Bragagnoli, A.; 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, 398, 27–40.
  36. Cunningham, D.; Allum, W.H.; Stenning, S.P.; Thompson, J.N.; Van de Velde, C.J.H.; Nicolson, M.; Scarffe, J.H.; Lofts, F.J.; Falk, S.J.; Iveson, T.J.; et al. Perioperative Chemotherapy versus Surgery Alone for Resectable Gastroesophageal Cancer. N. Engl. J. Med. 2006, 355, 11–20.
  37. Bang, Y.-J.; Kim, Y.-W.; Yang, H.-K.; Chung, H.C.; Park, Y.-K.; Lee, K.H.; Lee, K.-W.; Kim, Y.H.; Noh, S.-I.; Cho, J.Y.; et al. Adjuvant Capecitabine and Oxaliplatin for Gastric Cancer after D2 Gastrectomy (CLASSIC): A Phase 3 Open-Label, Randomised Controlled Trial. Lancet 2012, 379, 315–321.
  38. Smyth, E.C.; Wotherspoon, A.; Peckitt, C.; Gonzalez, D.; Hulkki-Wilson, S.; Eltahir, Z.; Fassan, M.; Rugge, M.; Valeri, N.; Okines, A.; et al. Mismatch Repair Deficiency, Microsatellite Instability, and Survival: An Exploratory Analysis of the Medical Research Council Adjuvant Gastric Infusional Chemotherapy (MAGIC) Trial. JAMA Oncol. 2017, 3, 1197–1203.
  39. Choi, Y.Y.; Kim, H.; Shin, S.-J.; Kim, H.Y.; Lee, J.; Yang, H.-K.; Kim, W.H.; Kim, Y.-W.; Kook, M.-C.; Park, Y.K.; et al. Microsatellite Instability and Programmed Cell Death-Ligand 1 Expression in Stage II/III Gastric Cancer: Post Hoc Analysis of the CLASSIC Randomized Controlled Study. Ann. Surg. 2019, 270, 309–316.
  40. Pietrantonio, F.; Miceli, R.; Raimondi, A.; Kim, Y.W.; Kang, W.K.; Langley, R.E.; Choi, Y.Y.; Kim, K.-M.; Nankivell, M.G.; Morano, F.; et al. Individual Patient Data Meta-Analysis of the Value of Microsatellite Instability As a Biomarker in Gastric Cancer. J. Clin. Oncol. 2019, 37, 3392–3400.
  41. Zhao, L.; Liu, Y.; Zhang, S.; Wei, L.; Cheng, H.; Wang, J.; Wang, J. Impacts and Mechanisms of Metabolic Reprogramming of Tumor Microenvironment for Immunotherapy in Gastric Cancer. Cell Death Dis. 2022, 13, 378.
  42. Oya, Y.; Hayakawa, Y.; Koike, K. Tumor Microenvironment in Gastric Cancers. Cancer Sci. 2020, 111, 2696–2707.
  43. Amedei, A.; Della Bella, C.; Silvestri, E.; Prisco, D.; D’Elios, M.M. T Cells in Gastric Cancer: Friends or Foes. J. Immunol. Res. 2012, 2012, 690571.
  44. Itahashi, K.; Irie, T.; Nishikawa, H. Regulatory T-Cell Development in the Tumor Microenvironment. Eur. J. Immunol. 2022, 52, 1216–1227.
  45. Zou, W.; Chen, L. Inhibitory B7-Family Molecules in the Tumour Microenvironment. Nat. Rev. Immunol. 2008, 8, 467–477.
  46. Gandhi, L.; Rodríguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; De Angelis, F.; Domine, M.; Clingan, P.; Hochmair, M.J.; Powell, S.F.; et al. Pembrolizumab plus Chemotherapy in Metastatic Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 2078–2092.
  47. Tallón de Lara, P.; Cecconi, V.; Hiltbrunner, S.; Yagita, H.; Friess, M.; Bode, B.; Opitz, I.; Vrugt, B.; Weder, W.; Stolzmann, P.; et al. Gemcitabine Synergizes with Immune Checkpoint Inhibitors and Overcomes Resistance in a Preclinical Model and Mesothelioma PatientsGemcitabine Synergizes with Immune Checkpoint Inhibitors. Clin. Cancer Res. 2018, 24, 6345–6354.
  48. Motzer, R.J.; Penkov, K.; Haanen, J.; Rini, B.; Albiges, L.; Campbell, M.T.; Venugopal, B.; Kollmannsberger, C.; Negrier, S.; Uemura, M.; et al. Avelumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2019, 380, 1103–1115.
  49. Emens, L.A.; Middleton, G. The Interplay of Immunotherapy and Chemotherapy: Harnessing Potential Synergies. Cancer Immunol. Res 2015, 3, 436–443.
  50. Huo, G.; Liu, W.; Chen, P. Efficacy of PD-1/PD-L1 Inhibitors in Gastric or Gastro-Oesophageal Junction Cancer Based on Clinical Characteristics: A Meta-Analysis. BMC Cancer 2023, 23, 143.
  51. Qing, Y.; Li, Q.; Ren, T.; Xia, W.; Peng, Y.; Liu, G.-L.; Luo, H.; Yang, Y.-X.; Dai, X.-Y.; Zhou, S.-F.; et al. Upregulation of PD-L1 and APE1 is Associated with Tumorigenesis and Poor Prognosis of Gastric Cancer. Drug Des. Devel. Ther. 2015, 9, 901–909.
  52. Shitara, K.; Van Cutsem, E.; Bang, Y.-J.; Fuchs, C.; Wyrwicz, L.; Lee, K.-W.; Kudaba, I.; Garrido, M.; Chung, H.C.; Lee, J.; et al. Efficacy and Safety of Pembrolizumab or Pembrolizumab plus Chemotherapy vs Chemotherapy Alone for Patients with First-Line, Advanced Gastric Cancer: The KEYNOTE-062 Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1571–1580.
  53. Chao, J.; Fuchs, C.S.; Shitara, K.; Tabernero, J.; Muro, K.; Van Cutsem, E.; Bang, Y.-J.; De Vita, F.; Landers, G.; Yen, C.-J.; et al. Assessment of Pembrolizumab Therapy for the Treatment of Microsatellite Instability-High Gastric or Gastroesophageal Junction Cancer Among Patients in the KEYNOTE-059, KEYNOTE-061, and KEYNOTE-062 Clinical Trials. JAMA Oncol. 2021, 7, 895–902.
  54. Dubois, M.; Liscia, N.; Brunetti, O.; Ziranu, P.; Lai, E.; Argentiero, A.; Mazza, E.; Cascinu, S.; Silvestris, N.; Casadei-Gardini, A.; et al. The Role of Immune Checkpoint Inhibitors in the Treatment Sequence of Advanced Gastric or Gastro-Esophageal Junction Cancer: A Systematic Review and Meta-Analysis of Randomized Trials. Crit. Rev. Oncol. Hematol. 2022, 173, 103674.
  55. André, T.; Tougeron, D.; Piessen, G.; de la Fouchardière, C.; Louvet, C.; Adenis, A.; Jary, M.; Tournigand, C.; Aparicio, T.; Desrame, J.; et al. Neoadjuvant Nivolumab Plus Ipilimumab and Adjuvant Nivolumab in Localized Deficient Mismatch Repair/Microsatellite Instability-High Gastric or Esophagogastric Junction Adenocarcinoma: The GERCOR NEONIPIGA Phase II Study. J. Clin. Oncol. 2023, 41, 255–265.
  56. Becker, K.; Mueller, J.D.; Schulmacher, C.; Ott, K.; Fink, U.; Busch, R.; Böttcher, K.; Siewert, J.R.; Höfler, H. Histomorphology and Grading of Regression in Gastric Carcinoma Treated with Neoadjuvant Chemotherapy. Cancer 2003, 98, 1521–1530.
  57. Pietrantonio, F.; Raimondi, A.; Lonardi, S.; Murgioni, S.; Cardellino, G.G.; Tamberi, S.; Strippoli, A.; Palermo, F.; Prisciandaro, M.; Randon, G.; et al. INFINITY: A Multicentre, Single-Arm, Multi-Cohort, Phase II Trial of Tremelimumab and Durvalumab as Neoadjuvant Treatment of Patients with Microsatellite Instability-High (MSI) Resectable Gastric or Gastroesophageal Junction Adenocarcinoma (GAC/GEJAC). J. Clin. Orthod. 2023, 41, 358.
  58. Ajani, J.A.; D’Amico, T.A.; Bentrem, D.J.; Chao, J.; Cooke, D.; Corvera, C.; Das, P.; Enzinger, P.C.; Enzler, T.; Fanta, P.; et al. Gastric Cancer, Version 2.2022, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Canc. Netw. 2022, 20, 167–192.
  59. Kelly, R.J.; Ajani, J.A.; Kuzdzal, J.; Zander, T.; Van Cutsem, E.; Piessen, G.; Mendez, G.; Feliciano, J.; Motoyama, S.; Lièvre, A.; et al. Adjuvant Nivolumab in Resected Esophageal or Gastroesophageal Junction Cancer. N. Engl. J. Med. 2021, 384, 1191–1203.
  60. Xu, J.; Jiang, H.; Pan, Y.; Gu, K.; Cang, S.; Han, L.; Shu, Y.; Li, J.; Zhao, J.; Pan, H.; et al. Abstract CT078: First-Line Treatment with Sintilimab (Sin) vs. Placebo in Combination with Chemotherapy (Chemo) in Patients (Pts) with Unresectable Gastric or Gastroesophageal Junction (G/GEJ) Cancer: Final Overall Survival (OS) Results from the Randomized, Phase III ORIENT-16 Trial. Cancer Res. 2023, 83, CT078.
  61. Moehler, M.H.; Kato, K.; Arkenau, H.-T.; Oh, D.-Y.; Tabernero, J.; Cruz-Correa, M.; Wang, H.; Xu, H.; Li, J.; Yang, S.; et al. Rationale 305: Phase 3 Study of Tislelizumab plus Chemotherapy vs Placebo plus Chemotherapy as First-Line Treatment (1L) of Advanced Gastric or Gastroesophageal Junction Adenocarcinoma (GC/GEJC). J. Clin. Orthod. 2023, 41, 286.
  62. Kang, Y.-K.; Chen, L.-T.; Ryu, M.-H.; Oh, D.-Y.; Oh, S.C.; Chung, H.C.; Lee, K.-W.; Omori, T.; Shitara, K.; Sakuramoto, S.; et al. Nivolumab plus Chemotherapy versus Placebo plus Chemotherapy in Patients with HER2-Negative, Untreated, Unresectable Advanced or Recurrent Gastric or Gastro-Oesophageal Junction Cancer (ATTRACTION-4): A Randomised, Multicentre, Double-Blind, Placebo-Controlled, Phase 3 Trial. Lancet Oncol. 2022, 23, 234–247.
  63. Raufi, A.G.; Lee, S.; May, M.; Portillo, A.D.; Sender, N.; Ana, S.S.; Gautier, K.; Alouani, E.; Park, H.; Oberstein, P.; et al. Abstract CT009: Phase II Trial of Perioperative Pembrolizumab plus Capecitabine and Oxaliplatin Followed by Adjuvant Pembrolizumab for Resectable Gastric and Gastroesophageal Junction (GC/GEJ) Adenocarcinoma. Cancer Res. 2022, 82, CT009.
  64. Al-Batran, S.-E.; Lorenzen, S.; Thuss-Patience, P.C.; Homann, N.; Schenk, M.; Lindig, U.; Heuer, V.; Kretzschmar, A.; Goekkurt, E.; Haag, G.M.; et al. Surgical and Pathological Outcome, and Pathological Regression, in Patients Receiving Perioperative Atezolizumab in Combination with FLOT Chemotherapy versus FLOT Alone for Resectable Esophagogastric Adenocarcinoma: Interim Results from DANTE, a Randomized, Multicenter, Phase IIb Trial of the FLOT-AIO German Gastric Cancer Group and Swiss SAKK. J. Clin. Orthod. 2022, 40, 4003.
  65. Bang, Y.-J.; Van Cutsem, E.; Fuchs, C.S.; Ohtsu, A.; Tabernero, J.; Ilson, D.H.; Hyung, W.J.; Strong, V.E.; Goetze, T.O.; Yoshikawa, T.; et al. KEYNOTE-585: Phase III Study of Perioperative Chemotherapy with or without Pembrolizumab for Gastric Cancer. Future Oncol. 2019, 15, 943–952.
  66. Janjigian, Y.Y.; Van Cutsem, E.; Muro, K.; Wainberg, Z.; Al-Batran, S.-E.; Hyung, W.J.; Molena, D.; Marcovitz, M.; Ruscica, D.; Robbins, S.H.; et al. MATTERHORN: Phase III Study of Durvalumab plus FLOT Chemotherapy in Resectable Gastric/Gastroesophageal Junction Cancer. Future Oncol. 2022, 18, 2465–2473.
  67. Hollebecque, A.; Wainberg, Z.A.; Ajani, J.A.; Marshall, J.; Cunningham, D.; Ou, S.-H.I.; Lutzky, J.; Jamal, R.; Curigliano, G.; Gutierrez, M.; et al. Safety and Clinical Activity of Durvalumab Monotherapy in Patients with Gastroesophageal Cancers. J. Clin. Orthod. 2018, 36, 4032.
  68. Kelly, R.J.; Lee, J.; Bang, Y.-J.; Almhanna, K.; Blum-Murphy, M.; Catenacci, D.V.T.; Chung, H.C.; Wainberg, Z.A.; Gibson, M.K.; Lee, K.-W.; et al. Safety and Efficacy of Durvalumab and Tremelimumab Alone or in Combination in Patients with Advanced Gastric and Gastroesophageal Junction Adenocarcinoma. Clin. Cancer Res. 2020, 26, 846–854.
  69. Terashima, M.; Kang, Y.-K.; Kim, Y.-W.; Boku, N.; Chung, H.C.C.; Chen, J.-S.; Ji, J.; Yeh, T.-S.; Chen, L.-T.; Ryu, M.-H.; et al. ATTRACTION-5: A Phase 3 Study of Nivolumab plus Chemotherapy as Postoperative Adjuvant Treatment for Pathological Stage III (PStage III) Gastric or Gastroesophageal Junction (G/GEJ) Cancer. J. Clin. Orthod. 2023, 41, 4000.
  70. Yuan, S.; Nie, R.-C.; Jin, Y.; Liang, C.-C.; Jian, R.; Li, Y.-F.; Qiu, H.; Wang, W.; Chen, S.; Zhang, D.-S.; et al. Perioperative PD-1 Antibody Toripalimab plus SOX or XELOX Chemotherapy versus SOX or XELOX Alone for Locally Advanced Gastric or Gastro-Oesophageal Junction Cancer: Results from a Prospective, Randomized, Open-Label, Phase II Trial. J. Clin. Orthod. 2023, 41, 4001.
More
This entry is offline, you can click here to edit this entry!
Video Production Service