Published online Dec 27, 2024. doi: 10.4240/wjgs.v16.i12.3881
Revised: September 24, 2024
Accepted: October 22, 2024
Published online: December 27, 2024
Processing time: 133 Days and 21.6 Hours
Gastric cancer (GC) metastasis is the main cause of poor prognosis for GC pa
Core Tip: Immunotherapy has gradually begun to be applied in the treatment of gastric cancer (GC) metastasis. Some studies have shown that liver metastases of advanced GC have a reduced immune response rate due to the presence of hepatic immune tolerance mechanisms. Therefore, the mechanism and efficacy of immunotherapy applied in other metastases are discussed. Previous studies have shown that immunotherapy has the best efficacy for lung metastasis and the worst efficacy for peritoneal metastasis, but it has a good safety profile and is promising for use in all GC metastases.
- Citation: Xie FF, Qian ST, Zhao HY, Liu QS. Therapeutic efficacy of immunotherapy for gastric cancer metastasis. World J Gastrointest Surg 2024; 16(12): 3881-3886
- URL: https://www.wjgnet.com/1948-9366/full/v16/i12/3881.htm
- DOI: https://dx.doi.org/10.4240/wjgs.v16.i12.3881
Gastric cancer (GC) is a global health challenge, ranking as the fifth most common malignant tumor worldwide[1,2]. Over one million people are diagnosed with GC each year worldwide, and the five-year survival rate for GC patients is less than 40%[3]. Early GC (EGC) is similar to the common symptoms of gastritis, when it develops in the late stage that obvious symptoms are detected. However, EGC accounts for 50%-80% of all GC cases[4].Common complications of GC include gastrointestinal bleeding, perforation, pyloric obstruction, and metastasis[5]. Metastasis accounts for the majority of cancer-related deaths[6]. Common metastases from GC including those in the liver, lung, peritoneum, bone and pancreas are relatively rare[7,8]. Current treatment methods for metastasis include surgery, chemotherapy, immunotherapy, targeted therapy, conversion therapy, radiation therapy, and transhepatic arterial chemoembolization (TACE)[9]. Immunotherapy, an emerging anti-cancer strategy, recognizes and removes tumor cells by activating and enhancing the body's own immune system. In recent years, immunotherapy has shown significant efficacy in a variety of post-metastatic solid tumors, including malignant melanoma[10]. Research on immunotherapy for GC, non-small cell lung cancer, and breast cancer, including non-specific enhancer therapy, immune cell therapy, tumor vaccines, oncolytic viruses, and immune checkpoint inhibitors has been conducted[11-13]. These immunotherapies work by reactivating and maintaining the tumor immune cycle, restoring the body's normal anti-tumor immune response, and controlling and eliminating tumors[14]. In recent years, relevant studies have identified several potential predictive biomarkers for prognosis and immunotherapeutic efficacy across various tumors, such as SLC35A2, LMNB2, glycosylation-related genes, and EPHB2. The newly identified biomarkers offer valuable insights into the tumor dynamics and progression of various cancers, including GC. They also serve as predictive markers for tumor prognosis and immunotherapy for cancer metastasis[15-18]. The papers entitled “Analysis of the impact of immunotherapy efficacy and safety in patients with GC and liver metastasis"[19] and “Gastric cancer liver metastasis will reduce the efficacy of immunotherapy”[20], both in the World Journal of Gastrointestinal Surgery, aroused our interest. Due to their observations that liver metastasis from GC reduces the efficacy of immunotherapy, but few studies have examined whether secondary tumors after GC metastasis affect the efficacy of immunotherapy. Therefore, this paper aims to elucidate the immunological mechanisms underlying GC metastasis and to evaluate the therapeutic efficacy of immunotherapy for various metastatic manifestations of GC, thereby offering more precise and effective treatment options for patients with metastatic GC.
High infiltration of immunosuppressive cells, such as regulatory T cells, myeloid-derived suppressor cells and tumor-associated macrophages, is common in GC metastases. These cells inhibit the function of effector T cells by secreting inhibitory cytokines (e.g., IL-10, TGF-β) and expressing immune checkpoint molecules (e.g., PD-L1), thereby promoting tumor cell metastasis and survival[21]. GC liver metastases can siphon activated CD8+ T cells into the somatic circulation. This siphoning function leads to ''immune deserts'' causing a decrease in the effectiveness of immunotherapy[22].
The PD-1/PD-L1 pathway is one of the most important immune checkpoints in GC metastasis. Studies have shown that the expression level of PD-L1 in metastatic GC tissues is significantly higher than that in primary foci, which may be one of the important mechanisms by which tumor cells evade immune surveillance. In addition, other immune checkpoint molecules, such as CTLA-4, TIM-3 and LAG-3, also play important roles in GC metastasis[23].
Tumor neoantigens are tumor-specific antigens produced as a result of genetic mutations that can be recognized by the immune system. Tumor-associated antigens loaded DC vaccines in cancer treatment have been extensively investigated in clinical trials. Activation of neoantigen-specific CD4+ and CD8+ T cells increases the frequency of neoantigen-specific T cell clones. One patient with metastatic GC who received Neo-MoDC vaccination co-mediated with ICI and is now improving and has survived for > 25 months[24].
During GC metastasis, the balance between cytokines and immune cells (e.g., VEGF, IL-10, TGF-β) is disrupted, creating a microenvironment that favors tumor growth and metastasis. This imbalance in the cytokine network also affects the function and differentiation of immune cells[25].
The liver is the most common site of GC metastasis, and the leading cause of death. The 1-year survival rate for patients with liver metastases is 15.1%[26]. Immunotherapies for GC liver metastases include the following: Systematic immunotherapy: For patients with unresectable GC with liver metastases, immune checkpoint inhibitors alone or in combination with chemotherapy may offer new treatment options. The KEYNOTE-061 study demonstrated that pembrolizumab shows good efficacy in patients with metastatic GC who had failed prior therapy, including those with liver metastases[27]. Local immunotherapy: Transhepatic arterial infusion of immunotherapeutic agents or combined embolization may improve local treatment outcomes. Combining PD-1 inhibitors with TACE may improve the prognosis of patients with liver metastases from GC[28].
GC metastasizes to the lungs or chest cavity, manifesting as multiple lung metastases, carcinomatous lymphadenitis, or carcinomatous pleurisy[29]. Immunotherapy for lung metastases is similar to that for liver metastases. The use of immune checkpoint inhibitors such as PD-1 inhibitors (e.g., pembrolizumab) and PD-L1 inhibitors (e.g., nabulizumab) enhances the ability of T cells to kill cancer cells by blocking the interactions between PD-1 and PD-L1 and relieving the tumor's suppression of immune cells in tomors[30]. Cellular immunotherapy can be used as a kind of auxiliary treatment in patients with GC lung metastasis, collecting the patient's own immune cells, culturing and expanding them in vitro then infusing them back into the patient's body, to enhance the body's immune response to the tumor. It is used in combination with chemotherapy and radiotherapy to improve therapeutic effects[31].
Peritoneal metastasis (PM) is a common site of advanced GC (AGC) metastasis, and is the most difficult type of meta
The use of pembrolizumab as neoadjuvant therapy significantly improves the rate of complete pathological remission in patients with locally AGC metastases. In patients who have developed lymph node metastases, the use of postoperative immunotherapy may reduce the risk of recurrence. The results from the CheckMate 577 trial revealed that the use of nabulizumab as an adjuvant therapy significantly improved disease-free survival in patients with adenocarcinoma of the esophagus or gastroesophageal junction treated with neoadjuvant radiotherapy and surgery[35]. A case report of a reduction in previously metabolically active lymph nodes in the left clavicular region, abdominal cavity, retroperitoneum, and bilateral parietal iliac vessels following chemotherapy in combination with tirilizumab treatment was followed by maintenance of tirilizumab monotherapy for up to 2 years, with no evidence of recurrence during the concluding follow-up period[36].
The remission rate of AGC patients treated with PD-1 inhibitors in combination with chemotherapy ranges from 50% to 65%[37,38]. In the previous two articles, we reported that liver metastases reduce the effectiveness of immunotherapy. Patients with lung cancer presenting with bone metastases have lower PD-L1 expression, which tends to affect the tumor immune microenvironment and decrease the effectiveness of immunotherapy. Although this study addresses lung cancer bone metastases, a similar situation may also occur in GC lung metastases[39]. Compared with hepatic metastases, where the effect on T-cell apoptosis leads to systemic immunosuppression, pulmonary metastases are much smaller, and are therefore considered to have a lesser impact on immunotherapy. Another study showed that in the PD-L1 Low-expression cohort, there was a significant correlation between higher response rates to PD-1 inhibitor combination chemotherapy in patients with non-diffuse disease, GEJ cancers, distant lymph node metastases, hepatic metastases, non-peritoneal metastases, and HER2-positive patients[40]. According to the post hoc analysis of ATTRACTION-2, PM negatively impacted the therapeutic efficacy of navulizumab in GC salvage therapy. Similarly, in this study, PM was an independent risk factor for poor PFS and OS in the total population and in the PD-L1 Low-expression group receiving PD-1 inhibitors plus chemotherapy as first-line treatment.
Immunotherapy in patients with lung metastases has the best prognosis. Although liver metastasis reduces the effectiveness of immunotherapy in patients, combination with chemotherapy can still significantly improve the long-term prognosis with good safety. The efficacy of treatment for the PM of GC remains to be studied. The literature concerning lymph node metastasis mixed and needs a lot of experimental verification. However, immunotherapy for the treatment of GC metastasis has shown good application prospects and has brought new hope to patients with GC metastasis. Identifying the optimal patient population for immunotherapy remains a challenge. Biomarkers such as PD-L1 expression, MSI-H status and tumor mutational burden are being evaluated to better select patients most likely to benefit. Further research is also needed to optimize combination strategies, including the timing and sequencing of immunotherapy with chemotherapy and other targeted therapies. Advances in new technologies, including ctDNA monitoring and artificial intelligence-based digital pathology, are expected to improve the precision of immunotherapy. By un
We gratefully acknowledge the kind cooperation of all authors in the preparation of this paper.
1. | Thrift AP, El-Serag HB. Burden of Gastric Cancer. Clin Gastroenterol Hepatol. 2020;18:534-542. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 432] [Cited by in F6Publishing: 827] [Article Influence: 206.8] [Reference Citation Analysis (1)] |
2. | Yang WJ, Zhao HP, Yu Y, Wang JH, Guo L, Liu JY, Pu J, Lv J. Updates on global epidemiology, risk and prognostic factors of gastric cancer. World J Gastroenterol. 2023;29:2452-2468. [PubMed] [DOI] [Cited in This Article: ] [Cited by in CrossRef: 72] [Cited by in F6Publishing: 73] [Article Influence: 73.0] [Reference Citation Analysis (7)] |
3. | Sun Y, Yu W, Guan W, Cai L, Qiao M, Zheng L, Jiang R, Wang R, Wang L. Integrated assessment of PD-L1 expression and molecular classification facilitates therapy selection and prognosis prediction in gastric cancer. Cancer Manag Res. 2019;11:6397-6410. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 11] [Cited by in F6Publishing: 11] [Article Influence: 2.2] [Reference Citation Analysis (0)] |
4. | Ajani JA, D'Amico TA, Bentrem DJ, Chao J, Cooke D, Corvera C, Das P, Enzinger PC, Enzler T, Fanta P, Farjah F, Gerdes H, Gibson MK, Hochwald S, Hofstetter WL, Ilson DH, Keswani RN, Kim S, Kleinberg LR, Klempner SJ, Lacy J, Ly QP, Matkowskyj KA, McNamara M, Mulcahy MF, Outlaw D, Park H, Perry KA, Pimiento J, Poultsides GA, Reznik S, Roses RE, Strong VE, Su S, Wang HL, Wiesner G, Willett CG, Yakoub D, Yoon H, McMillian N, Pluchino LA. Gastric Cancer, Version 2.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2022;20:167-192. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 41] [Cited by in F6Publishing: 699] [Article Influence: 349.5] [Reference Citation Analysis (0)] |
5. | Seeneevassen L, Bessède E, Mégraud F, Lehours P, Dubus P, Varon C. Gastric Cancer: Advances in Carcinogenesis Research and New Therapeutic Strategies. Int J Mol Sci. 2021;22. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 23] [Cited by in F6Publishing: 78] [Article Influence: 26.0] [Reference Citation Analysis (0)] |
6. | Fares J, Fares MY, Khachfe HH, Salhab HA, Fares Y. Molecular principles of metastasis: a hallmark of cancer revisited. Signal Transduct Target Ther. 2020;5:28. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 854] [Cited by in F6Publishing: 1111] [Article Influence: 277.8] [Reference Citation Analysis (0)] |
7. | Crippa S, Angelini C, Mussi C, Bonardi C, Romano F, Sartori P, Uggeri F, Bovo G. Surgical treatment of metastatic tumors to the pancreas: a single center experience and review of the literature. World J Surg. 2006;30:1536-1542. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 117] [Cited by in F6Publishing: 114] [Article Influence: 6.3] [Reference Citation Analysis (0)] |
8. | Xiaobin C, Zhaojun X, Tao L, Tianzeng D, Xuemei H, Fan Z, Chunyin H, Jianqiang H, Chen L. Analysis of Related Risk Factors and Prognostic Factors of Gastric Cancer with Bone Metastasis: A SEER-Based Study. J Immunol Res. 2022;2022:3251051. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1] [Cited by in F6Publishing: 12] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
9. | Li T, He Y, Zhong Q, Yu J, Chen X. Advances in Treatment Models of Advanced Gastric Cancer. Technol Cancer Res Treat. 2022;21:15330338221090353. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 3] [Cited by in F6Publishing: 1] [Article Influence: 0.5] [Reference Citation Analysis (0)] |
10. | Zaremba A, Zimmer L, Griewank KG, Ugurel S, Roesch A, Schadendorf D, Livingstone E. [Immunotherapy for malignant melanoma]. Internist (Berl). 2020;61:669-675. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 5] [Cited by in F6Publishing: 8] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
11. | Guan WL, He Y, Xu RH. Gastric cancer treatment: recent progress and future perspectives. J Hematol Oncol. 2023;16:57. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 179] [Reference Citation Analysis (0)] |
12. | Reck M, Remon J, Hellmann MD. First-Line Immunotherapy for Non-Small-Cell Lung Cancer. J Clin Oncol. 2022;40:586-597. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 25] [Cited by in F6Publishing: 391] [Article Influence: 195.5] [Reference Citation Analysis (0)] |
13. | Ye F, Dewanjee S, Li Y, Jha NK, Chen ZS, Kumar A, Vishakha, Behl T, Jha SK, Tang H. Advancements in clinical aspects of targeted therapy and immunotherapy in breast cancer. Mol Cancer. 2023;22:105. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 125] [Cited by in F6Publishing: 135] [Article Influence: 135.0] [Reference Citation Analysis (0)] |
14. | Zhang Z, Liu N, Sun M. Research Progress of Immunotherapy for Gastric Cancer. Technol Cancer Res Treat. 2023;22:15330338221150555. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 2] [Reference Citation Analysis (0)] |
15. | Xu S, Chen X, Fang J, Chu H, Fang S, Zeng L, Ma H, Zhang T, Chen Y, Wang T, Zhang X, Shen T, Zheng Y, Xu D, Lu Z, Pan Y, Liu Y. Comprehensive analysis of 33 human cancers reveals clinical implications and immunotherapeutic value of the solute carrier family 35 member A2. Front Immunol. 2023;14:1155182. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 3] [Reference Citation Analysis (0)] |
16. | Xu S, Liu Y, Ma H, Fang S, Wei S, Li X, Lu Z, Zheng Y, Liu T, Zhu X, Xu D, Pan Y. A Novel Signature Integrated of Immunoglobulin, Glycosylation and Anti-Viral Genes to Predict Prognosis for Breast Cancer. Front Genet. 2022;13:834731. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 4] [Reference Citation Analysis (0)] |
17. | Xu S, Lu Z. The role of LMNB2 as a diagnostic and prognostic biomarker in lung adenocarcinoma. Asian J Surg. 2024;. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
18. | Xu S, Zheng Y, Ye M, Shen T, Zhang D, Li Z, Lu Z. Comprehensive pan-cancer analysis reveals EPHB2 is a novel predictive biomarker for prognosis and immunotherapy response. BMC Cancer. 2024;24:1064. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
19. | Liu K, Wu CX, Liang H, Wang T, Zhang JY, Wang XT. Analysis of the impact of immunotherapy efficacy and safety in patients with gastric cancer and liver metastasis. World J Gastrointest Surg. 2024;16:700-709. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
20. | Wang L, Liu SS, Zhang SM, Chen XQ, Huang T, Tian R, Zhao YQ, Chen Z, Xianba CR. Gastric cancer liver metastasis will reduce the efficacy of immunotherapy. World J Gastrointest Surg. 2024;16:2760-2764. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
21. | Zheng P, Luo Q, Wang W, Li J, Wang T, Wang P, Chen L, Zhang P, Chen H, Liu Y, Dong P, Xie G, Ma Y, Jiang L, Yuan X, Shen L. Tumor-associated macrophages-derived exosomes promote the migration of gastric cancer cells by transfer of functional Apolipoprotein E. Cell Death Dis. 2018;9:434. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 166] [Cited by in F6Publishing: 252] [Article Influence: 42.0] [Reference Citation Analysis (0)] |
22. | Du Y, Lin Y, Gan L, Wang S, Chen S, Li C, Hou S, Hu B, Wang B, Ye Y, Shen Z. Potential crosstalk between SPP1 + TAMs and CD8 + exhausted T cells promotes an immunosuppressive environment in gastric metastatic cancer. J Transl Med. 2024;22:158. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 3] [Reference Citation Analysis (0)] |
23. | Cheng R, Li B, Wang H, Zeng Y. Immune checkpoint inhibitors and cellular immunotherapy for advanced gastric, gastroesophageal cancer: a long pathway. Clin Transl Oncol. 2023;25:3122-3138. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 4] [Reference Citation Analysis (0)] |
24. | Guo Z, Yuan Y, Chen C, Lin J, Ma Q, Liu G, Gao Y, Huang Y, Chen L, Chen LZ, Huang YF, Wang H, Li B, Chen Y, Zhang X. Durable complete response to neoantigen-loaded dendritic-cell vaccine following anti-PD-1 therapy in metastatic gastric cancer. NPJ Precis Oncol. 2022;6:34. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1] [Cited by in F6Publishing: 34] [Article Influence: 17.0] [Reference Citation Analysis (37)] |
25. | Park HS, Kwon WS, Park S, Jo E, Lim SJ, Lee CK, Lee JB, Jung M, Kim HS, Beom SH, Park JY, Kim TS, Chung HC, Rha SY. Comprehensive immune profiling and immune-monitoring using body fluid of patients with metastatic gastric cancer. J Immunother Cancer. 2019;7:268. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 12] [Cited by in F6Publishing: 22] [Article Influence: 4.4] [Reference Citation Analysis (0)] |
26. | Horn SR, Stoltzfus KC, Lehrer EJ, Dawson LA, Tchelebi L, Gusani NJ, Sharma NK, Chen H, Trifiletti DM, Zaorsky NG. Epidemiology of liver metastases. Cancer Epidemiol. 2020;67:101760. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 45] [Cited by in F6Publishing: 126] [Article Influence: 31.5] [Reference Citation Analysis (0)] |
27. | Shitara K, Van Cutsem E, Bang YJ, Fuchs C, Wyrwicz L, Lee KW, Kudaba I, Garrido M, Chung HC, Lee J, Castro HR, Mansoor W, Braghiroli MI, Karaseva N, Caglevic C, Villanueva L, Goekkurt E, Satake H, Enzinger P, Alsina M, Benson A, Chao J, Ko AH, Wainberg ZA, Kher U, Shah S, Kang SP, Tabernero J. 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. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 583] [Cited by in F6Publishing: 727] [Article Influence: 181.8] [Reference Citation Analysis (0)] |
28. | Xu J, Liu H, Ni G, Huang Y, Huang Y, Liang H, Ni Y, Huang Q, Yang Z. Clinical efficacy of PD-1 inhibitor combined with radiotherapy in a multi-drug resistant patient with liver metastasis from gastric cancer. Front Surg. 2023;10:1101294. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1] [Reference Citation Analysis (0)] |
29. | Shigenobu T, Ohtsuka T, Hanawa R, Sakamaki H, Yoshizu A, Tajima A. Prognostic Impact of Visceral Pleural Invasion in Resected Solitary Lung Metastases from Gastric Cancer. Ann Thorac Cardiovasc Surg. 2023;29:279-286. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
30. | Tu J, Xu H, Ma L, Li C, Qin W, Chen X, Yi M, Sun L, Liu B, Yuan X. Nintedanib enhances the efficacy of PD-L1 blockade by upregulating MHC-I and PD-L1 expression in tumor cells. Theranostics. 2022;12:747-766. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 4] [Cited by in F6Publishing: 23] [Article Influence: 11.5] [Reference Citation Analysis (0)] |
31. | Kan N, Imamura M. [Loco-regional immunotherapy with OK-432 and cultured autologous lymphocytes for patients with metastatic cancer]. Hum Cell. 1993;6:100-105. [PubMed] [Cited in This Article: ] |
32. | Arita T, Ichikawa D, Konishi H, Komatsu S, Shiozaki A, Ogino S, Fujita Y, Hiramoto H, Hamada J, Shoda K, Kosuga T, Fujiwara H, Okamoto K, Otsuji E. Tumor exosome-mediated promotion of adhesion to mesothelial cells in gastric cancer cells. Oncotarget. 2016;7:56855-56863. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 38] [Cited by in F6Publishing: 46] [Article Influence: 7.7] [Reference Citation Analysis (0)] |
33. | Chen C, Jung A, Yang A, Monroy I, Zhang Z, Chaurasiya S, Deshpande S, Priceman S, Fong Y, Park AK, Woo Y. Chimeric Antigen Receptor-T Cell and Oncolytic Viral Therapies for Gastric Cancer and Peritoneal Carcinomatosis of Gastric Origin: Path to Improving Combination Strategies. Cancers (Basel). 2023;15. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
34. | Yarema R, Mielko J, Fetsych T, Ohorchak M, Skorzewska M, Rawicz-Pruszyński K, Mashukov A, Maksimovsky V, Jastrzębski T, Polkowski W, Gyrya P, Kovalchuk Y, Safiyan V, Karelin I, Kopetskiy V, Kolesnik O, Kondratskiy Y, Paskonis M. Hyperthermic intraperitoneal chemotherapy (HIPEC) in combined treatment of locally advanced and intraperitonealy disseminated gastric cancer: A retrospective cooperative Central-Eastern European study. Cancer Med. 2019;8:2877-2885. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 25] [Cited by in F6Publishing: 38] [Article Influence: 7.6] [Reference Citation Analysis (0)] |
35. | Lin Y, Liang HW, Liu Y, Pan XB. Nivolumab adjuvant therapy for esophageal cancer: a review based on subgroup analysis of CheckMate 577 trial. Front Immunol. 2023;14:1264912. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 1] [Reference Citation Analysis (0)] |
36. | Zhu Z, Dai PL, Han S, Qiu E, Wang Y, Li Z. Complete remission in a patient with metastatic gastric cancer receiving tislelizumab combined with chemotherapy: a case report. Front Oncol. 2023;13:1147636. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
37. | Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, Wyrwicz L, Yamaguchi K, Skoczylas T, Campos Bragagnoli A, Liu T, Schenker M, Yanez P, Tehfe M, Kowalyszyn R, Karamouzis MV, Bruges R, Zander T, Pazo-Cid R, Hitre E, Feeney K, Cleary JM, Poulart V, Cullen D, Lei M, Xiao H, Kondo K, Li M, Ajani JA. 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. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1201] [Cited by in F6Publishing: 1514] [Article Influence: 504.7] [Reference Citation Analysis (0)] |
38. | Xu J, Jiang H, Pan Y, Gu K, Cang S, Han L, Shu Y, Li J, Zhao J, Pan H, Luo S, Qin Y, Guo Q, Bai Y, Ling Y, Yang J, Yan Z, Yang L, Tang Y, He Y, Zhang L, Liang X, Niu Z, Zhang J, Mao Y, Guo Y, Peng B, Li Z, Liu Y, Wang Y, Zhou H; ORIENT-16 Investigators. Sintilimab Plus Chemotherapy for Unresectable Gastric or Gastroesophageal Junction Cancer: The ORIENT-16 Randomized Clinical Trial. JAMA. 2023;330:2064-2074. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 15] [Cited by in F6Publishing: 44] [Article Influence: 44.0] [Reference Citation Analysis (0)] |
39. | Zhu YJ, Chang XS, Zhou R, Chen YD, Ma HC, Xiao ZZ, Qu X, Liu YH, Liu LR, Li Y, Yu YY, Zhang HB. Bone metastasis attenuates efficacy of immune checkpoint inhibitors and displays "cold" immune characteristics in Non-small cell lung cancer. Lung Cancer. 2022;166:189-196. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 2] [Cited by in F6Publishing: 2] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
40. | Sun YT, Lu SX, Lai MY, Yang X, Guan WL, Yang LQ, Li YH, Wang FH, Yang DJ, Qiu MZ. Clinical outcomes and biomarker exploration of first-line PD-1 inhibitors plus chemotherapy in patients with low PD-L1-expressing of gastric or gastroesophageal junction adenocarcinoma. Cancer Immunol Immunother. 2024;73:144. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1] [Reference Citation Analysis (0)] |