101
|
Pan Y, Ma Y, Dai G. Prognostic value of a novel myeloid-to-lymphoid ratio biomarker in advanced gastric cancer. Clin Transl Oncol 2024:10.1007/s12094-024-03612-3. [PMID: 39141277 DOI: 10.1007/s12094-024-03612-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/09/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Currently, immune checkpoint inhibitors (ICIs) have excellent performance in the clinical treatment of advanced gastric cancer (AGC). However, precisely selecting AGC patients who can benefit from immunotherapy is an urgent difficulty. In this study, we investigated the immunoprognostic role of myeloid-to-lymphocyte ratio (M:L) in AGC patients. METHODS We collected information on 268 AGC patients who were hospitalized in the Department of Medical Oncology of PLA General Hospital from December 2014 to May 2021. The patients were divided into low M: L group (< 3.76) and high M:L group (≥ 3.76). Survival differences between different M: L level groups at baseline and after treatment were analyzed by methods such as Kaplan-Meier, Cox or Logistic regression model. RESULTS Progression free survival (PFS) (5.8 months vs. 3.4 months, p = 0.001) and overall survival (OS) (14.1 months vs. 9.0 months, p = 0.001) were significantly longer in the low M:L group than in the high M:L group. After analyses of Cox regression modeling it was concluded that M:L was an independent prognostic factor for PFS (HR 1.371 95%CI 1.057-1.777 p = 0.017) and OS (HR 1.352 95%CI 1.003-1.824 p = 0.048), respectively. Subsequent subgroup analyses performed across immunotherapy lines, regimens, PD-1 inhibitor agents, and age groups revealed a poorer prognosis in the high M:L group. Notably, an increase in the value of M:L after treatment significantly increased the risk of poor prognosis. CONCLUSIONS M:L ≥ 3.76 is associated with poor prognostic outcomes in AGC patients receiving immunotherapy and may be a predictive biomarker of prognosis. This result needs to be confirmed by larger prospective studies.
Collapse
Affiliation(s)
- Yuting Pan
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Medical Oncology, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
- Department of Medical Oncology, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yue Ma
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Medical Oncology, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
- Department of Medical Oncology, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Guanghai Dai
- Medical School of Chinese PLA, Beijing, 100853, China.
- Department of Medical Oncology, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.
- Department of Medical Oncology, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
102
|
Lim SH, Saluja A, Vickers S, Hong JY, Kim ST, Lavania S, Pandey S, Gupta VK, Velagapudi MR, Lee J. The safety and efficacy outcomes of Minnelide given alone or in combination with paclitaxel in advanced gastric cancer: A phase I trial. Cancer Lett 2024; 597:217041. [PMID: 38866072 DOI: 10.1016/j.canlet.2024.217041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
Minnelide is a water-soluble disodium salt variant of triptolide, an HSP70 inhibitor that can prevent tumor progression and induce apoptosis. Maximum tolerated dose (MTD), safety, and antitumor activity of Minnelide alone and its combination with paclitaxel were evaluated in this open-label, single-center, dose-escalation phase I study (NCT05566834) in patients who were previously treated for advanced gastric cancer (AGC). Minnelide was administered orally using a 3 + 3 dose-escalation design as monotherapy (Regimen A), and in combination with paclitaxel (Regimen B & C). Our results show that no patients experienced dose limiting toxicity (DLT) in the combination group (Regimen B& C) while 2 patients experienced DLT from the Regimen A group (n = 11) (Minnelide 1.5 mg). The MTD was Minnelide 1.25 mg once daily for 21days Q4 weeks as monotherapy. The most common Grade ≥3 AEs were neutropenia (19.4 %) and abdominal pain (11.1 %). In Regimen C, 71.5 % achieved either a partial response or a stable disease with the median PFS of 4.5 months, and the median OS of 10.7 months. The combination of Minnelide plus paclitaxel as salvage treatment in AGC patients showed meaningful clinical activity with a manageable safety profile. Based on these encouraging results, a phase II study is being initiated to test the effectiveness of the combination regimen in patients with advanced gastric cancer.
Collapse
Affiliation(s)
- Sung Hee Lim
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Ashok Saluja
- Minneamrita Therapeutics LLC, Tampa, FL, 33647, USA
| | | | - Jung Yong Hong
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Seung Tae Kim
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Shweta Lavania
- Department of Surgery, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Somnath Pandey
- Department of Surgery, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Vineet K Gupta
- Department of Surgery, University of Miami School of Medicine, Miami, FL, 33136, USA
| | | | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea.
| |
Collapse
|
103
|
Chong X, Madeti Y, Cai J, Li W, Cong L, Lu J, Mo L, Liu H, He S, Yu C, Zhou Z, Wang B, Cao Y, Wang Z, Shen L, Wang Y, Zhang X. Recent developments in immunotherapy for gastrointestinal tract cancers. J Hematol Oncol 2024; 17:65. [PMID: 39123202 PMCID: PMC11316403 DOI: 10.1186/s13045-024-01578-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
The past few decades have witnessed the rise of immunotherapy for Gastrointestinal (GI) tract cancers. The role of immune checkpoint inhibitors (ICIs), particularly programmed death protein 1 (PD-1) and PD ligand-1 antibodies, has become increasingly pivotal in the treatment of advanced and perioperative GI tract cancers. Currently, anti-PD-1 plus chemotherapy is considered as first-line regimen for unselected advanced gastric/gastroesophageal junction adenocarcinoma (G/GEJC), mismatch repair deficient (dMMR)/microsatellite instability-high (MSI-H) colorectal cancer (CRC), and advanced esophageal cancer (EC). In addition, the encouraging performance of claudin18.2-redirected chimeric antigen receptor T-cell (CAR-T) therapy in later-line GI tract cancers brings new hope for cell therapy in solid tumour treatment. Nevertheless, immunotherapy for GI tumour remains yet precise, and researchers are dedicated to further maximising and optimising the efficacy. This review summarises the important research, latest progress, and future directions of immunotherapy for GI tract cancers including EC, G/GEJC, and CRC.
Collapse
Affiliation(s)
- Xiaoyi Chong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Yelizhati Madeti
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Jieyuan Cai
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Wenfei Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Lin Cong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Jialin Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Liyang Mo
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Huizhen Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Siyi He
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Chao Yu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Zhiruo Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Boya Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Yanshuo Cao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yakun Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
104
|
Marashi A, Hasany S, Moghimi S, Kiani R, Mehran Asl S, Dareghlou YA, Lorestani P, Varmazyar S, Jafari F, Ataeian S, Naghavi K, Sajjadi SM, Haratian N, Alinezhad A, Azhdarimoghaddam A, Sadat Rafiei SK, Anar MA. Targeting gut-microbiota for gastric cancer treatment: a systematic review. Front Med (Lausanne) 2024; 11:1412709. [PMID: 39170038 PMCID: PMC11337614 DOI: 10.3389/fmed.2024.1412709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024] Open
Abstract
Background Preclinical research has identified the mechanisms via which bacteria influence cancer treatment outcomes. Clinical studies have demonstrated the potential to modify the microbiome in cancer treatment. Herein, we systematically analyze how gut microorganisms interact with chemotherapy and immune checkpoint inhibitors, specifically focusing on how gut bacteria affect the pharmacokinetics and pharmacodynamics of cancer treatment. Method This study searched Web of Science, Scopus, and PubMed until August 2023. Studies were screened by their title and abstract using the Rayyan intelligent tool for systematic reviews. Quality assessment of studies was done using the JBI critical appraisal tool. Result Alterations in the gut microbiome are associated with gastric cancer and precancerous lesions. These alterations include reduced microbial alpha diversity, increased bacterial overgrowth, and decreased richness and evenness of gastric bacteria. Helicobacter pylori infection is associated with reduced richness and evenness of gastric bacteria, while eradication only partially restores microbial diversity. The gut microbiome also affects the response to cancer treatments, with higher abundances of Lactobacillus associated with better response to anti-PD-1/PD-L1 immunotherapy and more prolonged progression-free survival. Antibiotic-induced gut microbiota dysbiosis can reduce the anti-tumor efficacy of 5-Fluorouracil treatment, while probiotics did not significantly enhance it. A probiotic combination containing Bifidobacterium infantis, Lactobacillus acidophilus, Enterococcus faecalis, and Bacillus cereus can reduce inflammation, enhance immunity, and restore a healthier gut microbial balance in gastric cancer patients after partial gastrectomy. Conclusion Probiotics and targeted interventions to modulate the gut microbiome have shown promising results in cancer prevention and treatment efficacy.Systematic review registration: https://osf.io/6vcjp.
Collapse
Affiliation(s)
- Amir Marashi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saina Hasany
- Student Research Committee, Islamic Azad University Tehran Medical Sciences, Tehran, Iran
| | - Sadra Moghimi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Kiani
- Student Research Committee, Islamic Azad University Tehran Medical Sciences, Mashhad, Iran
| | - Sina Mehran Asl
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Islamic Azad University Tehran Medical Sciences, Tehran, Iran
| | | | - Parsa Lorestani
- School of Medicine, Shahroud Azad University of Medical Sciences, Shahroud, Iran
| | - Shirin Varmazyar
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Alborz, Iran
| | - Fatemeh Jafari
- School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shakiba Ataeian
- School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kiana Naghavi
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Negar Haratian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arman Alinezhad
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Mahsa Asadi Anar
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
105
|
Kang YK, Terashima M, Kim YW, Boku N, Chung HC, Chen JS, Ji J, Yeh TS, Chen LT, Ryu MH, Kim JG, Omori T, Rha SY, Kim TY, Ryu KW, Sakuramoto S, Nishida Y, Fukushima N, Yamada T, Bai LY, Hirashima Y, Hagihara S, Nakada T, Sasako M. Adjuvant nivolumab plus chemotherapy versus placebo plus chemotherapy for stage III gastric or gastro-oesophageal junction cancer after gastrectomy with D2 or more extensive lymph-node dissection (ATTRACTION-5): a randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Gastroenterol Hepatol 2024; 9:705-717. [PMID: 38906161 DOI: 10.1016/s2468-1253(24)00156-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND In Asia, adjuvant chemotherapy after gastrectomy with D2 or more extensive lymph-node dissection is standard treatment for people with pathological stage III gastric or gastro-oesophageal junction (GEJ) cancer. We aimed to assess the efficacy and safety of adjuvant nivolumab plus chemotherapy versus placebo plus chemotherapy administered in this setting. METHODS ATTRACTION-5 was a randomised, multicentre, double-blind, placebo-controlled, phase 3 trial conducted at 96 hospitals in Japan, South Korea, Taiwan, and China. Eligible patients were aged between 20 years and 80 years with histologically confirmed pathological stage IIIA-C gastric or GEJ adenocarcinoma after gastrectomy with D2 or more extensive lymph-node dissection, with an Eastern Cooperative Oncology Group (ECOG) performance status score of 0 or 1 and available tumour tissue for PD-L1 expression analysis. Patients were randomly assigned (1:1) to receive either nivolumab plus chemotherapy or placebo plus chemotherapy via an interactive web-response system with block sizes of four. Investigational treatment, either nivolumab 360 mg or placebo, was administered intravenously for 30 min once every 3 weeks. Adjuvant chemotherapy was administered as either tegafur-gimeracil-oteracil (S-1) at an initial dose of 40 mg/m2 per dose orally twice per day for 28 consecutive days, followed by 14 days off per cycle, or capecitabine plus oxaliplatin consisting of an initial dose of intravenous oxaliplatin 130 mg/m2 for 2 h every 21 days and capecitabine 1000 mg/m2 per dose orally twice per day for 14 consecutive days, followed by 7 days off treatment. The primary endpoint was relapse-free survival by central assessment. The intention-to-treat population, consisting of all randomly assigned patients, was used for analysis of efficacy endpoints. The safety population, defined as patients who received at least one dose of trial drug, was used for analysis of safety endpoints. This trial is registered with ClinicalTrials.gov (NCT03006705) and is closed. FINDINGS Between Feb 1, 2017, and Aug 15, 2019, 755 patients were randomly assigned to receive either adjuvant nivolumab plus chemotherapy (n=377) or adjuvant placebo plus chemotherapy (n=378). 267 (71%) of 377 patients in the nivolumab group and 263 (70%) of 378 patients in the placebo group were male; 110 (29%) of 377 patients in the nivolumab group and 115 (31%) of 378 patients in the placebo group were female. 745 patients received assigned treatment (371 in the nivolumab plus chemotherapy group; 374 in the placebo plus chemotherapy group), which was the safety population. Median time from first dose to data cutoff was 49·1 months (IQR 43·1-56·7). 3-year relapse-free survival was 68·4% (95% CI 63·0-73·2) in the nivolumab plus chemotherapy group and 65·3% (59·9-70·2) in the placebo plus chemotherapy group; the hazard ratio for relapse-free survival was 0·90 (95·72% CI 0·69-1·18; p=0·44). Treatment-related adverse events occurred in 366 (99%) of 371 patients in the nivolumab plus chemotherapy group and 364 (98%) of 374 patients in the placebo plus chemotherapy group. Discontinuation due to adverse events was more frequent in the nivolumab plus chemotherapy group (34 [9%] of 371 patients) than the placebo plus chemotherapy group (13 [4%] of 374 patients). The most common treatment-related adverse events were decreased appetite, nausea, diarrhoea, neutrophil count decreased, and peripheral sensory neuropathy. INTERPRETATION The results of this trial do not support the addition of nivolumab to postoperative adjuvant therapy for patients with untreated, locally advanced, resectable gastric or GEJ cancer. FUNDING Ono Pharmaceutical and Bristol Myers Squibb.
Collapse
Affiliation(s)
- Yoon-Koo Kang
- Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea
| | | | - Young-Woo Kim
- Surgical Department, Center for Gastric Cancer, National Cancer Center, Goyang, South Korea
| | - Narikazu Boku
- Department of Oncology and General Medicine, Institute of Medical Science, University of Tokyo Hospital, Tokyo, Japan
| | - Hyun Cheol Chung
- Yonsei Cancer Center, Yonsei University Health System, Seoul, South Korea
| | - Jen-Shi Chen
- Division of Hematology and Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Chang Gung University, Taoyuan City, Taiwan
| | - Jiafu Ji
- Department of Gastrointestinal Surgery, Beijing Cancer Hospital, Beijing, China
| | - Ta-Sen Yeh
- Department of Surgery, Linkou Chang Gung Memorial Hospital, Chang Gung University, Taoyuan City, Taiwan
| | - Li-Tzong Chen
- Centre for Cancer Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan; Department of Internal Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan City, Taiwan
| | - Min-Hee Ryu
- Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Jong Gwang Kim
- Department of Hemato-Oncology, Chilgok Hospital, Kyungpook National University, Daegu, South Korea
| | - Takeshi Omori
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Sun Young Rha
- Yonsei Cancer Center, Yonsei University Health System, Seoul, South Korea
| | - Tae Yong Kim
- Oncology Department, Seoul National University Hospital, Seoul, South Korea
| | - Keun Won Ryu
- Surgical Department, Center for Gastric Cancer, National Cancer Center, Goyang, South Korea
| | - Shinichi Sakuramoto
- Department of Gastrointestinal Surgery, International Medical Center, Saitama Medical University, Saitama, Japan
| | - Yasunori Nishida
- Department of Gastrointestinal Surgery and Medical Oncology, Keiyukai Sapporo Hospital, Sapporo, Japan
| | - Norimasa Fukushima
- Department of Surgery, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Takanobu Yamada
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Li-Yuan Bai
- Division of Hematology and Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | | | | | | | - Mitsuru Sasako
- Department of Surgery, Yodogawa Christian Hospital, Osaka, Japan.
| |
Collapse
|
106
|
Qi C, Liu C, Gong J, Liu D, Wang X, Zhang P, Qin Y, Ge S, Zhang M, Peng Z, Zhou J, Lu Z, Lu M, Cao Y, Yuan J, Wang Y, Wang Z, Xue R, Peng X, Wang Y, Yuan D, Li J, Zhang X, Shen L. Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial final results. Nat Med 2024; 30:2224-2234. [PMID: 38830992 DOI: 10.1038/s41591-024-03037-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/30/2024] [Indexed: 06/05/2024]
Abstract
Claudin18.2 (CLDN18.2) is highly expressed with the development of various malignant tumors, especially gastrointestinal cancers, and is emerging as a new target for cancer treatment. Satricabtagene autoleucel (satri-cel)/CT041 is an autologous chimeric antigen receptor (CAR) T cell targeting CLDN18.2, and the interim results of the CT041-CG4006 trial were reported in June 2022. Here we present the final results of this single-arm, open-label, phase 1 trial, which evaluated the safety and efficacy of satri-cel in patients with CLDN18.2-positive advanced gastrointestinal cancers. This trial included a dose-escalation stage (n = 15) and a dose-expansion stage in four different cohorts (total n = 83): cohort 1, satri-cel monotherapy in 61 patients with standard chemotherapy-refractory gastrointestinal cancers; cohort 2, satri-cel plus anti-PD-1 therapy in 15 patients with standard chemotherapy-refractory gastrointestinal cancers; cohort 3, satri-cel as sequential treatment after first-line therapy in five patients with gastrointestinal cancers; and cohort 4, satri-cel monotherapy in two patients with anti-CLDN18.2 monoclonal antibody-refractory gastric cancer. The primary endpoint was safety; secondary endpoints included efficacy, pharmacokinetics and immunogenicity. A total of 98 patients received satri-cel infusion, among whom 89 were dosed with 2.5 × 108, six with 3.75 × 108 and three with 5.0 × 108 CAR T cells. Median follow-up was 32.4 months (95% confidence interval (CI): 27.3, 36.5) since apheresis. No dose-limiting toxicities, treatment-related deaths or immune effector cell-associated neurotoxicity syndrome were reported. Cytokine release syndrome occurred in 96.9% of patients, all classified as grade 1-2. Gastric mucosal injuries were identified in eight (8.2%) patients. The overall response rate and disease control rate in all 98 patients were 38.8% and 91.8%, respectively, and the median progression-free survival and overall survival were 4.4 months (95% CI: 3.7, 6.6) and 8.8 months (95% CI: 7.1, 10.2), respectively. Satri-cel demonstrates therapeutic potential with a manageable safety profile in patients with CLDN18.2-positive advanced gastrointestinal cancer. ClinicalTrials.gov identifier: NCT03874897 .
Collapse
Affiliation(s)
- Changsong Qi
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Chang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jifang Gong
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Dan Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xicheng Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Panpan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Sai Ge
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Miao Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhi Peng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jun Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhihao Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ming Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanshuo Cao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiajia Yuan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yakun Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhenghang Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ran Xue
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | | | - Yumeng Wang
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | | | - Jian Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Xiaotian Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Lin Shen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
107
|
Tasaki Y, Ito N, Mimura Y, Sugiyama Y, Ogawa R, Shimura T, Nakamura M, Kawakita D, Hamamoto S, Uemura T, Yokota K, Iida M, Odagiri K, Kimura Y, Hotta Y, Komatsu H, Okuda K, Niimi A, Yasui T, Iwasaki S, Morita A, Kataoka H, Takiguchi S, Furukawa-Hibi Y. Real-world data on efficacy/safety and economic impact of nivolumab administered every 2 and 4 weeks among Japanese patients. Asia Pac J Clin Oncol 2024; 20:515-521. [PMID: 38682421 DOI: 10.1111/ajco.14073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 03/06/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024]
Abstract
AIM A new treatment interval for nivolumab administration at 480 mg every 4 weeks, in addition to 240 mg every 2 weeks, was approved in Japan in 2020. Using model-based evaluation, it was speculated that the effects or safety of nivolumab do not differ between the two treatment intervals; however, real-world data on nivolumab efficacy, safety, and economic impact are lacking. Accordingly, we aimed to examine the effects of nivolumab treatment intervals (2 weeks vs. 4 weeks) in terms of efficacy, safety, and economic impact in Japanese patients with cancer. METHODS We retrospectively analyzed 126 patients treated with nivolumab. The patients were divided into two groups depending on whether they received nivolumab at 240 mg every 2 weeks (2-week group) or 480 mg every 4 weeks (4-week group). RESULTS Efficacy results found no significant difference between the 4- and 2-week groups considering median overall survival (p = 0.70) and median progression-free survival (p = 0.57). The incidence of any grade and ≥ grade 3 immune-related adverse events did not differ between the 4-week and 2-week groups (any grade, p = 0.13; ≥ grade 3, p = 0.36). Excluding drug costs, the 4-week group had significantly lower medical costs than the 2-week group (2-week vs. 4-week: mean, 94,659 JPY [679.0 USD] vs. 58,737 JPY [421.3 USD]; p < 0.05). CONCLUSION Collectively, our findings suggest that nivolumab 480 mg every 4 weeks may be more effective than nivolumab 240 mg every 2 weeks in terms of economic impact.
Collapse
Affiliation(s)
- Yoshihiko Tasaki
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Nanami Ito
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoshihisa Mimura
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yosuke Sugiyama
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ryo Ogawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takaya Shimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Motoki Nakamura
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Daisuke Kawakita
- Department of Otorhinolaryngology, Head and Neck Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shuzo Hamamoto
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takehiro Uemura
- Department of Respiratory Medicine, Allergy, and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Keisuke Yokota
- Department of Thoracic and Pediatric Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Moeko Iida
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kunihiro Odagiri
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuka Kimura
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuji Hotta
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hirokazu Komatsu
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Katsuhiro Okuda
- Department of Thoracic and Pediatric Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akio Niimi
- Department of Respiratory Medicine, Allergy, and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takahiro Yasui
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shinichi Iwasaki
- Department of Otorhinolaryngology, Head and Neck Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akimichi Morita
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiromi Kataoka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoko Furukawa-Hibi
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
108
|
Wang SY, Yang XQ, Wang YX, Shen A, Liang CC, Huang RJ, Cheng UH, Jian R, An N, Xiao YL, Wang LS, Zhao Y, Lin C, Wang CP, Yuan ZP, Yuan SQ. Overexpression of COX7A1 Promotes the Resistance of Gastric Cancer to Oxaliplatin and Weakens the Efficacy of Immunotherapy. J Transl Med 2024; 104:102090. [PMID: 38830579 DOI: 10.1016/j.labinv.2024.102090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/09/2024] [Accepted: 05/28/2024] [Indexed: 06/05/2024] Open
Abstract
Gastric cancer (GC) is one of the most common clinical malignant tumors worldwide, with high morbidity and mortality. Presently, the overall response rate to immunotherapy is low, and current methods for predicting the prognosis of GC are not optimal. Therefore, novel biomarkers with accuracy, efficiency, stability, performance ratio, and wide clinical application are needed. Based on public data sets, the chemotherapy cohort and immunotherapy cohort from Sun Yat-sen University Cancer Center, a series of bioinformatics analyses, such as differential expression analysis, survival analysis, drug sensitivity prediction, enrichment analysis, tumor immune dysfunction and exclusion analysis, single-sample gene set enrichment analysis, stemness index calculation, and immune cell infiltration analysis, were performed for screening and preliminary exploration. Immunohistochemical staining and in vitro experiments were performed for further verification. Overexpression of COX7A1 promoted the resistance of GC cells to Oxaliplatin. COX7A1 may induce immune escape by regulating the number of fibroblasts and their cellular communication with immune cells. In summary, measuring the expression levels of COX7A1 in the clinic may be useful in predicting the prognosis of GC patients, the degree of chemotherapy resistance, and the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Si-Yu Wang
- Department of Oncology, The First People's Hospital of Yibin, Yibin, China
| | - Xian-Qi Yang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yu-Xin Wang
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, China
| | - Ao Shen
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng-Cai Liang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Run-Jie Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Un Hio Cheng
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rui Jian
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Nan An
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yu-Long Xiao
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Li-Shuai Wang
- Department of Oncology, The First People's Hospital of Yibin, Yibin, China
| | - Yin Zhao
- Department of Oncology, The First People's Hospital of Yibin, Yibin, China
| | - Chuan Lin
- Department of Oncology, The First People's Hospital of Yibin, Yibin, China
| | - Chang-Ping Wang
- Department of Oncology, The First People's Hospital of Yibin, Yibin, China
| | - Zhi-Ping Yuan
- Department of Oncology, The First People's Hospital of Yibin, Yibin, China
| | - Shu-Qiang Yuan
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
109
|
He P, Ma L, Xu B, Wang Y, Li X, Chen H, Li Y. Research progress and future directions of immune checkpoint inhibitor combination therapy in advanced gastric cancer. Ther Adv Med Oncol 2024; 16:17588359241266156. [PMID: 39091604 PMCID: PMC11292724 DOI: 10.1177/17588359241266156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 06/18/2024] [Indexed: 08/04/2024] Open
Abstract
In recent years, with the continuous development of molecular immunology, immune checkpoint inhibitors (ICIs) have also been widely used in the treatment of gastric cancer, but they still face some challenges: The first is that only some people can benefit, the second is the treatment-related adverse events (TRAEs) that occur during treatment, and the third is the emergence of varying degrees of drug resistance with long-term use. How to overcome these challenges, combined therapy based on ICIs has become one of the important strategies. This article summarizes the clinical application of ICIs combined with chemotherapy, targeted therapy, radiotherapy, photodynamic therapy, thermotherapy, immune adjuvant, and dual immunotherapy and discusses the mechanism, and also summarizes the advantages and disadvantages of the current combination modalities and the potential research value. The aim of this study is to provide more and more optimized combination regimen for ICI combined therapy in patients with advanced gastric cancer and to provide reference for clinical and scientific research.
Collapse
Affiliation(s)
- Puyi He
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, China
| | - Long Ma
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, China
| | - Bo Xu
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, China
| | - Yunpeng Wang
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, China
| | - Xiaomei Li
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, China
| | - Hao Chen
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, Lanzhou, China
- No. 82, Cuiyingmen, Chengguan, Lanzhou 730030, China
| | - Yumin Li
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, Lanzhou, China
- No. 82, Cuiyingmen, Chengguan, Lanzhou 730030, China
| |
Collapse
|
110
|
Wang H, Chen J, Gao W, Wu Y, Wang X, Lin F, Chen H, Wang Y, Jiang T, Pan Z, Gao X, Liu Q, Weng X, Yao N, Zhu Y, Wu R, Weng G, Lin X. Construction of a nomogram with IrAE and clinic character to predict the survival of advanced G/GEJ adenocarcinoma patients undergoing anti-PD-1 treatment. Front Immunol 2024; 15:1432281. [PMID: 39114652 PMCID: PMC11303212 DOI: 10.3389/fimmu.2024.1432281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
Objective This study aimed to develop and validate a survival prediction model and nomogram to predict survival in patients with advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma undergoing treatment with anti-programmed cell death 1 receptor (PD-1). This model incorporates immune-related adverse events (irAEs) alongside common clinical characteristics as predictive factors. Method A dataset comprising 255 adult patients diagnosed with advanced G/GEJ adenocarcinoma was assembled. The irAEs affecting overall survival (OS) to a significant degree were identified and integrated as a candidate variable, together with 12 other candidate variables. These included gender, age, Eastern cooperative oncology group performance status (ECOG PS) score, tumor stage, human epidermal growth factor receptor 2 (HER2) expression status, presence of peritoneal and liver metastases, year and line of anti-PD-1 treatment, neutrophil-to-lymphocyte ratio (NLR), controlling nutritional status (CONUT) score, and Charlson comorbidity index (CCI). To mitigate timing bias related to irAEs, landmark analysis was employed. Variable selection was performed using the least absolute shrinkage and selection operator (LASSO) regression to pinpoint significant predictors, and the variance inflation factor was applied to address multicollinearity. Subsequently, a Cox regression analysis utilizing the forward likelihood ratio method was conducted to develop a survival prediction model, excluding variables that failed to satisfy the proportional hazards (PH) assumption. The model was developed using the entire dataset, then internally validated through bootstrap resampling and externally validated with a cohort from another Hospital. Furthermore, a nomogram was created to delineate the predictive model. Results After consolidating irAEs from the skin and endocrine systems into a single protective irAE category and applying landmark analysis, variable selection was conducted for the prognostic prediction model along with other candidate variables. The finalized model comprised seven variables: ECOG PS score, tumor stage, HER2 expression status in tumor tissue, first-line anti-PD-1 treatment, peritoneal metastasis, CONUT score, and protective irAE. The overall concordance index for the model was 0.66. Calibration analysis verified the model's accuracy in aligning predicted outcomes with actual results. Clinical decision curve analysis indicated that utilizing this model for treatment decisions could enhance the net benefit regarding 1- and 2-year survival rates for patients. Conclusion This study developed a prognostic prediction model by integrating common clinical characteristics of irAEs and G/GEJ adenocarcinoma. This model exhibits good clinical practicality and possesses accurate predictive ability for overall survival OS in patients with advanced G/GEJ adenocarcinoma.
Collapse
Affiliation(s)
- Han Wang
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jinhua Chen
- Follow-Up Center, Fujian Medical University Union Hospital, Fuzhou, China
| | - Wei Gao
- Departments of Internal Medicine-Oncology, Fujian Provincial Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
| | - Yilan Wu
- The School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xinli Wang
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Fangyu Lin
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Hao Chen
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yao Wang
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Tao Jiang
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhangchi Pan
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xinyan Gao
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qing Liu
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaojiao Weng
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Na Yao
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yingjiao Zhu
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Riping Wu
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Guizhen Weng
- Department of Oncology Nursing, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoyan Lin
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
111
|
Santero M, Meade AG, Selva A, Savall-Esteve O, Bracchiglione J, Macías I, Leache L, Cerdà P, Bonfill Cosp X. Utilising systematic reviews to assess potential overtreatment and claim for better evidence-based research: an analysis of anticancer drugs versus supportive care in advanced esophageal cancer. Syst Rev 2024; 13:186. [PMID: 39026378 PMCID: PMC11256491 DOI: 10.1186/s13643-024-02594-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Highlighting the identified gaps in evidence-based research concerning advanced esophageal cancer (EC) treatment and care, this review evaluates the efficacy and safety of anticancer drugs compared to supportive care for advanced EC patients, aiming to assess the appropriateness of usual treatments and identify the gaps that need to be filled with primary research. METHODS We searched (May 2022) MEDLINE, EMBASE, the Cochrane Central Register of Controlled Trials (CENTRAL), Epistemonikos, and trial registries (ClinicalTrials.gov and PROSPERO) for randomised controlled trials (RCTs) comparing anticancer drugs (chemotherapy, immunotherapy, or biological/targeted therapy) with supportive care in advanced EC. The results were summarised using GRADE summary of finding tables. RESULTS We included 15 RCTs. Most studies did not have a special focus on EC, did not detail the treatment lines in all patients, and did not evaluate all outcomes. Anticancer drugs may result in a slight increase in overall survival (OS) (HR 0.78; 95% CI 0.71, 0.86; MD 0.83 months) and better progression-free survival (PFS) (HR 0.56 95% CI 0.49, 0.64, MD 0.68 months), but also may increase toxicity (RR 1.37; 95% CI 1.13, 1.65), without a significant improvement in quality of life. The certainty of evidence was low or very low due to indirectness of results and lack of specific focus on EC in some studies. CONCLUSION RCTs on advanced EC lack specificity, detailed treatment line information, and evaluation of all relevant outcomes. Moreover, when they find any benefit, this is negligible. Therefore, the certainty to justify anticancer drug treatments instead of supportive care in advanced EC is low or very low, and this information should be actively shared with affected patients. More and better RCTs should be conducted to assess whether any old or new proposed treatment for advanced EC patients provides a better balance of benefits and harms than the supportive care. SYSTEMATIC REVIEW REGISTRATION The study protocol was registered in OSF ( https://doi.org/10.17605/OSF.IO/7CHX6 ) on 2022-03-29.
Collapse
Affiliation(s)
- Marilina Santero
- Universitat Autònoma Barcelona, Barcelona, Spain.
- Iberoamerican Cochrane Centre, Institut de Recerca Sant Pau (IR, SANT PAU), Barcelona, Spain.
| | - Adriana-Gabriela Meade
- Iberoamerican Cochrane Centre, Institut de Recerca Sant Pau (IR, SANT PAU), Barcelona, Spain
| | - Anna Selva
- Universitat Autònoma Barcelona, Barcelona, Spain
- Iberoamerican Cochrane Centre, Institut de Recerca Sant Pau (IR, SANT PAU), Barcelona, Spain
- Clinical Epidemiology and Cancer Screening, Parc Taulí Hospital Universitari, Institut d'Investigació I Innovació Parc Taulí (I3PT_CERCA), Sabadell, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Olga Savall-Esteve
- Universitat Autònoma Barcelona, Barcelona, Spain
- Iberoamerican Cochrane Centre, Institut de Recerca Sant Pau (IR, SANT PAU), Barcelona, Spain
| | - Javier Bracchiglione
- Universitat Autònoma Barcelona, Barcelona, Spain
- Iberoamerican Cochrane Centre, Institut de Recerca Sant Pau (IR, SANT PAU), Barcelona, Spain
- Interdisciplinary Centre for Health Studies (CIESAL), Universidad de Valparaíso, Viña del Mar, Chile
- CIBER de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Ismael Macías
- Servicio Oncología Médica, Hospital de Sabadell-Corporació Sanitària Parc Taulí, Sabadell, Barcelona, Spain
| | - Leire Leache
- Unit of Innovation and Organization, Navarre Health Service, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Paula Cerdà
- Universitat Autònoma Barcelona, Barcelona, Spain
- Iberoamerican Cochrane Centre, Institut de Recerca Sant Pau (IR, SANT PAU), Barcelona, Spain
- Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
| | - Xavier Bonfill Cosp
- Universitat Autònoma Barcelona, Barcelona, Spain
- Iberoamerican Cochrane Centre, Institut de Recerca Sant Pau (IR, SANT PAU), Barcelona, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
| |
Collapse
|
112
|
Pei X, Zhang SL, Qiu BQ, Zhang PF, Liu TS, Wang Y. Cancer Cell Secreted Legumain Promotes Gastric Cancer Resistance to Anti-PD-1 Immunotherapy by Enhancing Macrophage M2 Polarization. Pharmaceuticals (Basel) 2024; 17:951. [PMID: 39065799 PMCID: PMC11279811 DOI: 10.3390/ph17070951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/19/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
The interaction between cancer cells and immune cells plays critical roles in gastric cancer (GC) progression and immune evasion. Forced legumain (LGMN) is one of the characteristics correlated with poor prognosis in gastric cancer patients. However, the role of gastric-cancer-secreted LGMN (sLGMN) in modulating the tumor immune microenvironment and the biological effect on the immune evasion of gastric cancer remains unclear. In this study, we found that forced expression of sLGMN in gastric cancer serum correlates with increased M2 macrophage infiltration in GC tissues and predicted resistance to anti-PD-1 immunotherapy. Mechanistically, gastric cancer cells secrete LGMN via binding to cell surface Integrin αvβ3, then activate Integrin αvβ3/PI3K (Phosphatidylinositol-4,5-bisphosphate3-kinase)/AKT (serine/threonine kinase)/mTORC2 (mammalian target of rapamycin complex 2) signaling, promote metabolic reprogramming, and polarize macrophages from the M1 to the M2 phenotype. Either blocking LGMN, Integrin αv, or knocking out Integrin αv expression and abolishing the LGMN/Integrin αvβ3 interaction significantly inhibits metabolic reprogramming and polarizes macrophages from the M1 to the M2 phenotype. This study reveals a critical molecular crosstalk between gastric cancer cells and macrophages through the sLGMN/Integrinαvβ3/PI3K/AKT/mTORC2 axis in promoting gastric cancer immune evasion and resistance to anti-PD-1 immunotherapy, indicating that the sLGMN/Integrinαvβ3/PI3K/AKT/mTORC2 axis may act as a promising therapeutic target.
Collapse
Affiliation(s)
- Xu Pei
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (X.P.); (S.-L.Z.); (P.-F.Z.)
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330030, China;
| | - Shi-Long Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (X.P.); (S.-L.Z.); (P.-F.Z.)
| | - Bai-Quan Qiu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330030, China;
| | - Peng-Fei Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (X.P.); (S.-L.Z.); (P.-F.Z.)
- Department of Medical Oncology, Shanghai Geriatric Medical Center, Shanghai 201104, China
| | - Tian-Shu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (X.P.); (S.-L.Z.); (P.-F.Z.)
- Cancer Center, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, China
- Center of Evidence-Based Medicine, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, China
| | - Yan Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (X.P.); (S.-L.Z.); (P.-F.Z.)
- Cancer Center, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, China
- Center of Evidence-Based Medicine, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, China
- Shanghai Medical College, Zhongshan Hospital Immunotherapy Translational Research Center, Shanghai 200032, China
| |
Collapse
|
113
|
Tan S, Zheng Q, Zhang W, Zhou M, Xia C, Feng W. Prognostic value of inflammatory markers NLR, PLR, and LMR in gastric cancer patients treated with immune checkpoint inhibitors: a meta-analysis and systematic review. Front Immunol 2024; 15:1408700. [PMID: 39050856 PMCID: PMC11266030 DOI: 10.3389/fimmu.2024.1408700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) represent a groundbreaking approach to cancer therapy. Inflammatory markers such as the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and lymphocyte-to-monocyte ratio (LMR) have emerged as potential indicators strongly associated with tumor prognosis, albeit their prognostic significance remains contentious. The predictive value of NLR, PLR, LMR in patients with gastric cancer (GC) treated with ICIs has not been fully explored; therefore, we conducted a meta-analysis to examine the potential of inflammatory markers NLR, PLR, and LMR as survival predictors in this population. Methods A comprehensive search was conducted across PubMed, Embase, Web of Science, and Cochrane databases, with the search cut-off date set as March 2024. Hazard ratios (HR) and their corresponding 95% confidence intervals (CI) were calculated to assess the prognostic significance of NLR, PLR, and LMR for both progression-free survival (PFS) and overall survival (OS). Results Fifteen cohort studies involving 1336 gastric cancer patients were finally included in this meta-analysis. The results of the meta-analysis showed that high levels of NLR were associated with poorer OS and PFS in GC patients receiving ICIs, with combined HRs of OS [HR=2.01, 95%CI (1.72,2.34), P<0.01], and PFS PFS[HR=1.59, 95%CI (1.37,1.86), P<0.01], respectively; high levels of PLR were associated with poorer OS and PFS, and the combined HR was OS [HR=1.57, 95%CI (1.25,1.96), P<0.01], PFS [HR=1.52,95%CI (1.20, 1.94), P<0.01], respectively; and there was an association between elevated LMR and prolonged OS and PFS, and the combined HR was OS [HR=0.62, 95%CI (0.47,0.81), P<0.01], and PFS [HR=0.69, 95%CI (0.50,0.95), P<0.01]. Conclusion In gastric cancer (GC) patients treated with immune checkpoint inhibitors (ICIs), elevated neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) were associated with poorer overall survival (OS) and progression-free survival (PFS), while high lymphocyte-to-monocyte ratio (LMR) was linked to improved OS and PFS. Subgroup analyses suggested that NLR might be particularly pertinent to the prognosis of GC patients. In conclusion, the inflammatory markers NLR, PLR, and LMR serve as effective biomarkers for prognostic assessment in GC patients, offering valuable insights for therapeutic decision-making in the realm of GC immunotherapy. Prospective studies of high quality are eagerly awaited to validate these findings in the future. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/#myprospero, identifier CRD42024524321.
Collapse
Affiliation(s)
- Shufa Tan
- Shaanxi University of Traditional Chinese Medicine the First Clinical Medical College, Shaanxi, China
| | - Qin Zheng
- Fuling District Zhenxi Central Health Center, Inpatient Department, Chongqing, China
| | - Wei Zhang
- Shaanxi University of Traditional Chinese Medicine the First Clinical Medical College, Shaanxi, China
| | - Mi Zhou
- Physical Examination Center of Fuling Hospital Affiliated to Chongqing University, Chongqing, China
| | - Chunyan Xia
- Physical Examination Center of Fuling Hospital Affiliated to Chongqing University, Chongqing, China
| | - Wenzhe Feng
- Anorectal Department, Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Shaanxi, China
| |
Collapse
|
114
|
Tian Y, Yin Z, Zhang C, Li Z, Wang Y, Zhang K, Chen F, Dang Q. Differences in the risk of immune-related pneumonitis between PD-1 and PD-L1 inhibitors: a meta-analysis according to the new mirror-principle and PRISMA guidelines. Cancer Immunol Immunother 2024; 73:162. [PMID: 38953977 PMCID: PMC11219650 DOI: 10.1007/s00262-024-03736-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 05/15/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE To compare the risk of immune-associated pneumonitis between PD-1 and PD-L1 inhibitors, the meta-analysis was designed. METHOD The difference in risk of immune-associated pneumonitis between PD-1 and PD-L1 inhibitors was assessed by two different meta-analysis methods, the Mirror-pairing and the PRISMA guidelines. RESULTS A total of eighty-eight reports were used for meta-analysis, while thirty-two studies were used for the Mirror-pairing. Both PD-1 and PD-L1 inhibitors (used alone or combined with chemotherapy) increased the risk of developing immune-related pneumonitis (P < 0.00001; P < 0.00001). Based on indirect analyses results (subgroup analyses), the risk of PD-L1-induced pneumonitis was weaker than that of PD-1 inhibitors when the control group was chemotherapy (OR = 3.33 vs. 5.43) or placebo (OR = 2.53 vs. 3.19), while no obvious significant differences were found (P = 0.17; P = 0.53). For the Mirror-pairing-based meta-analysis, the risk of PD-1-induced pneumonitis was significantly higher than that of PD-L1 inhibitors (OR = 1.46, 95%CI [1.08, 1.98], I2 = 0%, Z = 2.47 (P = 0.01)). However, this difference was not significant, when they were combined with chemotherapy (OR = 1.05, 95%CI [0.68, 1.60], I2 = 38%, Z = 0.21 (P = 0.84)). CONCLUSION Both PD-1 and PD-L1 inhibitors increased the risk of immune-related pneumonitis, while the risk of PD-1-induced pneumonitis was significantly higher than that of PD-L1 inhibitors.
Collapse
Affiliation(s)
- Yuan Tian
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Huaiyin District, Jinan City, 250117, Shandong, People's Republic of China
- Radiotherapy Department, Shandong Second Provincial General Hospital, Shandong University, Jinan, 250299, Shandong, People's Republic of China
| | - Zongxiu Yin
- Department of Pulmonary and Critical Care Medicine, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, Shandong, People's Republic of China
| | - Chi Zhang
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, People's Republic of China
| | - Zhuoqi Li
- Radiotherapy Department, Shandong Second Provincial General Hospital, Shandong University, Jinan, 250299, Shandong, People's Republic of China
| | - Yuanyuan Wang
- Department of Oncology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250299, Shandong, People's Republic of China
| | - Kai Zhang
- General Surgery Department, Wen-Shang County People's Hospital, Wenshang, 272500, Shandong, People's Republic of China
| | - Feng Chen
- Department of Thoracic Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China
| | - Qi Dang
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Huaiyin District, Jinan City, 250117, Shandong, People's Republic of China.
| |
Collapse
|
115
|
Fong C, Patel B, Peckitt C, Bourmpaki E, Satchwell L, Cromarty S, Kidd S, von Loga K, Uhlik M, Begum R, Rana T, Waddell T, Darby S, Bradshaw A, Roques T, Morgan C, Rees C, Herbertson R, Das P, Thompson C, Hewish M, Petty R, Thistlethwaite F, Rao S, Starling N, Chau I, Cunningham D. Maintenance durvalumab after first-line chemotherapy in patients with HER2-negative advanced oesophago-gastric adenocarcinoma: results from the randomised PLATFORM study. ESMO Open 2024; 9:103622. [PMID: 39002179 PMCID: PMC11298822 DOI: 10.1016/j.esmoop.2024.103622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/26/2024] [Accepted: 05/31/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND PLAnning Treatment For Oesophago-gastric Cancer: a Randomised Maintenance Therapy Trial (PLATFORM) is an adaptive phase II study assessing the role of maintenance therapies in advanced oesophago-gastric (OG) adenocarcinoma. We evaluated the role of the anti-programmed death-ligand 1 (PD-L1) inhibitor durvalumab in these patients. PATIENTS AND METHODS Patients with human epidermal growth factor receptor 2-negative locally advanced or metastatic OG adenocarcinoma with disease control or response to 18 weeks of platinum-based first-line chemotherapy were randomised to active surveillance or maintenance durvalumab. The primary endpoint was progression-free survival (PFS). Safety was assessed in all patients who had commenced surveillance visits or received at least one dose of durvalumab. Exploratory survival analyses according to PD-L1 Combined Positive Score (CPS) and immune (biomarker-positive) or angiogenesis dominant (biomarker-negative) tumour microenvironment (TME) phenotypes were conducted. RESULTS Between March 2015 and April 2020, 205 patients were randomised to surveillance (n = 100) and durvalumab (n = 105). No significant differences were seen in PFS [hazard ratio (HR) 0.84, P = 0.13] and overall survival (OS; HR 0.98, P = 0.45) between surveillance and durvalumab. Five patients randomised to durvalumab demonstrated incremental radiological responses compared with none with surveillance. Treatment-related adverse events occurred in 77 (76.2%) durvalumab-assigned patients. A favourable effect in OS with durvalumab over surveillance in CPS ≥5 and immune biomarker-positive patients was observed compared with CPS <5 and biomarker-negative subgroups, respectively: CPS ≥5 versus <5: HR 0.63, 95% confidence interval (CI) 0.32-1.22 versus HR 0.93, 95% CI 0.44-1.96; biomarker-positive versus negative: HR 0.60, 95% CI 0.29-1.23 versus HR 0.84, 95% CI 0.42-1.65. CONCLUSION Maintenance durvalumab does not improve PFS in patients with OG adenocarcinoma who respond to first-line chemotherapy but induced incremental radiological responses in a subset of patients. TME characterisation could refine patient selection for anti-PD-L1 therapy above PD-L1 CPS alone.
Collapse
Affiliation(s)
- C Fong
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - B Patel
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - C Peckitt
- Research Data & Statistics Unit, Royal Marsden Clinical Trials Unit, Royal Marsden Hospital, London and Surrey
| | - E Bourmpaki
- Research Data & Statistics Unit, Royal Marsden Clinical Trials Unit, Royal Marsden Hospital, London and Surrey
| | - L Satchwell
- Research Data & Statistics Unit, Royal Marsden Clinical Trials Unit, Royal Marsden Hospital, London and Surrey
| | - S Cromarty
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - S Kidd
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - K von Loga
- Integrative Pathology Unit, Institute of Cancer Research, Sutton, UK
| | - M Uhlik
- Research and Biomarker Discovery, OncXerna Therapeutics, Inc., Waltham, USA
| | - R Begum
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - T Rana
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - T Waddell
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester
| | - S Darby
- Weston Park Cancer Centre, Sheffield
| | - A Bradshaw
- Cancer Services, The Freeman Hospital's Northern Centre for Cancer Care, Newcastle
| | - T Roques
- Department of Clinical Oncology, Norfolk and Norwich University Hospital, Norfolk & Norwich
| | - C Morgan
- Department of Clinical Oncology, Velindre Cancer Centre, Cardiff
| | - C Rees
- Cancer Care Division, Medical Oncology Department, University Hospital Southampton NHS Foundation Trust, Southampton
| | - R Herbertson
- Sussex Cancer Centre, Royal Sussex County Hospital, Brighton
| | - P Das
- Department of Oncology, University Hospitals of Derby and University of Nottingham School of Medicine
| | - C Thompson
- University Hospitals of Morecambe Bay NHS Foundation Trust, Kendal
| | - M Hewish
- Royal Surrey Cancer Centre, Royal Surrey Hospitals NHS Foundation Trust, Guildford
| | - R Petty
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee
| | - F Thistlethwaite
- Department of Medical Oncology, The Christie NHS Foundation Trust and Division of Cancer Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - S Rao
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - N Starling
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - I Chau
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - D Cunningham
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey.
| |
Collapse
|
116
|
Kono K, Nakajima S, Mimura K. Biomarker-oriented chemo-immunotherapy for advanced gastric cancer. Int J Clin Oncol 2024; 29:865-872. [PMID: 38647874 DOI: 10.1007/s10147-024-02525-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 04/25/2024]
Abstract
The biomarker-oriented chemo-immunotherapy is useful and promising in the development of new anticancer agents, since the responders can be enriched by selecting patients with biomarkers. Compared to colorectal and lung cancers, the development of biomarker-driven molecular-targeted therapeutics for gastric cancers has been straggled. However, several new biomarkers in gastric cancers have been discovered and clinical trials in enrichment design with certain biomarkers have been conducted. Therefore, there are currently several treatment options to treat gastric cancer patients based on individual biomarker-oriented strategies. In the present review, we describe the useful biomarkers in gastric cancer, with focusing on HER2, PD-L1, and Claudin18.2, in relation to their clinical significance and associated targeted agents.
Collapse
Affiliation(s)
- Koji Kono
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, 960-1295, Japan.
| | - Shotaro Nakajima
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, 960-1295, Japan
- Department of Multidisciplinary Treatment of Cancer and Regional Medical Support, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima City, Fukushima, 960-1295, Japan
| | - Kosaku Mimura
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, 960-1295, Japan
- Department of Blood Transfusion and Transplantation Immunology, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, 960-1295, Japan
| |
Collapse
|
117
|
Onishi M, Furuta M, Yoshioka E, Yamada T, Hama T, Furusawa K, Hayashi K, Inokuchi Y, Machida N, Furuse J, Maeda S. Complete response induced by nivolumab monotherapy in gastric neuroendocrine carcinoma: a case report. Int Cancer Conf J 2024; 13:319-324. [PMID: 38962039 PMCID: PMC11217240 DOI: 10.1007/s13691-024-00687-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/04/2024] [Indexed: 07/05/2024] Open
Abstract
No standard treatment has been established for gastric neuroendocrine carcinoma (G-NEC). We present the case of a patient with recurrent G-NEC who achieved a complete response (CR) with nivolumab. A woman in her 70 s, with no significant medical or family history of illness, underwent an upper gastrointestinal endoscopy, which revealed a Borrmann type 2 tumor in the gastric antrum. Malignant tumor cells were not detected in the endoscopic biopsy samples; however, a malignant gastric tumor was strongly suspected. Therefore, surgical resection was performed, and the tumor was pathologically diagnosed as a G-NEC with liver metastases. Adjuvant etoposide plus carboplatin was administered for four cycles, but recurrence in the liver was observed 5 months after the completion of adjuvant chemotherapy. Ramucirumab plus paclitaxel and irinotecan were introduced as second and third-line treatments. After these treatments, the mesenteric lymph node metastases expanded. Tumor mutation burden (TMB) was low (five mutations/megabase), and microsatellite instability remained stable. However, programmed death-ligand 1 Combined Positive Score (CPS) was ≥ 5 in the resected sample. Therefore, nivolumab monotherapy was introduced as a fourth-line treatment. The mesenteric lymph node metastases exhibited swelling 3 weeks after the initiation of nivolumab; however, they rapidly shrank, and CR was later achieved. Treatment with nivolumab is currently ongoing for 12 months. This is the first report of nivolumab monotherapy in a patient with G-NEC who showed pseudo-progression. Even in TMB-low and microsatellite stable cases, nivolumab may be a viable option for patients with G-NEC.
Collapse
Affiliation(s)
- Misa Onishi
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama-shi, Kanagawa 241-8515 Japan
| | - Mitsuhiro Furuta
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama-shi, Kanagawa 241-8515 Japan
| | - Emi Yoshioka
- Department of Pathology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama-shi, Kanagawa 241-8515 Japan
| | - Takanobu Yamada
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama-shi, Kanagawa 241-8515 Japan
| | - Takanori Hama
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama-shi, Kanagawa 241-8515 Japan
| | - Kyoko Furusawa
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama-shi, Kanagawa 241-8515 Japan
| | - Kei Hayashi
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama-shi, Kanagawa 241-8515 Japan
| | - Yasuhiro Inokuchi
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama-shi, Kanagawa 241-8515 Japan
| | - Nozomu Machida
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama-shi, Kanagawa 241-8515 Japan
| | - Junji Furuse
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama-shi, Kanagawa 241-8515 Japan
| | - Shin Maeda
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawaku, Yokohama, Kanagawa 236-0004 Japan
| |
Collapse
|
118
|
Mitrea DA, Froicu EM, Prenen H, Gambacorta MA, Span PN, Poortmans P. Combining immunotherapy and radiation therapy in gastrointestinal cancers: A review. Crit Rev Oncol Hematol 2024; 199:104381. [PMID: 38735504 DOI: 10.1016/j.critrevonc.2024.104381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 05/14/2024] Open
Abstract
INTRODUCTION AND PURPOSE With a significant global impact, treatment of gastrointestinal (GI) cancers still presents with challenges, despite current multimodality approaches in advanced stages. Clinical trials are expanding for checkpoint inhibition (ICI) combined with radiation therapy (RT). This review intends to offer a comprehensive image of the current data regarding the effectiveness of this association, and to reflect on possible directions to further optimize the results. RESULTS Several early phase studies demonstrated encouraging potential. However, translating preclinical outcomes to clinical settings proves challenging, especially in immunologically "cold" environments. GI cancers exhibit heterogeneity, requiring tailored approaches based on disease stage and patient characteristics. Current results, though promising, lack the power of evidence to influence the general practice. CONCLUSIONS Finding biomarkers for identifying or converting resistant cancers is essential for maximizing responses, moreover in this context strategic RT parameters need to be carefully considered. Our review emphasizes the significance of having a thorough grasp of how immunology, tumour biology, and treatment settings interact in order to propose novel research avenues and efficient GI cancer therapy.
Collapse
Affiliation(s)
- Diana A Mitrea
- Department of Radiation Oncology, Centre Antoine-Lacassagne, 33 Av. de Valombrose, Nice 06100, France.
| | - Eliza M Froicu
- Department of Medical Oncology, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
| | - Hans Prenen
- Department of Medical Oncology, Antwerp University Hospital, Edegem, Belgium
| | - Maria A Gambacorta
- Department of Radiation Oncology Fondazione Policlinico Universitario "A. Gemelli", Rome, Italy
| | - Paul N Span
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Philip Poortmans
- Department of Radiation Oncology, Iridium Netwerk, Wilrijk-Antwerp, Belgium; University of Antwerp, Faculty of Medicine and Health Sciences, Wilrijk-Antwerp, Belgium
| |
Collapse
|
119
|
Hiraizumi K, Honda C, Watanabe A, Nakao T, Midorikawa S, Abe H, Matsui N, Yamamoto T, Sakamoto T. Safety of nivolumab monotherapy in five cancer types: pooled analysis of post-marketing surveillance in Japan. Int J Clin Oncol 2024; 29:932-943. [PMID: 38844668 PMCID: PMC11196337 DOI: 10.1007/s10147-024-02515-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 03/13/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Nivolumab has been approved for treating ≥ 10 cancer types. However, there is limited information on the incidence of rare, but potentially serious, treatment-related adverse events (TRAEs), as well as notable TRAEs in patients with certain medical disorders or older patients in Japan. METHODS We performed pooled analyses of data from published post-marketing surveillance in Japan of nivolumab monotherapy for patients with malignant melanoma, non-small cell lung cancer, renal cell carcinoma, head and neck cancer, and gastric cancer to determine the frequencies of 20 categories of TRAEs of special interest overall and in patient groups with higher perceived safety risks (history of autoimmune disease, interstitial lung disease, tuberculosis, or hepatitis B/C; patients vaccinated during nivolumab treatment; and older patients [≥ 75 years]). RESULTS The overall population comprised 7421 patients treated with nivolumab. TRAEs were reported in 49.1% of patients, with grade ≥ 3 TRAEs in 16.7%. Endocrine disorders (14.4%), hepatobiliary disorders (10.9%), and interstitial lung disease (7.0%) were the three most common categories (any grade). The incidences of rare TRAEs with high risk of becoming serious, which occurred in < 1% of patients, were consistent with those in previous reports. The frequencies of TRAEs were not markedly increased in the specified patient groups relative to the overall population. CONCLUSION To our knowledge, this is the largest study examining the safety of nivolumab-treated patients in real-world clinical practice including rare but potentially serious TRAEs. We found no new signals in the safety of nivolumab among the patient groups relative to the overall population, and no additional safety measures are required in these groups. Trial registration UMIN000048892 (overall analysis), JapicCTI-163272 (melanoma), Japic-163271 (non-small cell lung cancer), JapicCTI-184071 (head and neck cancer), JapicCTI-184070 (gastric cancer), and JapicCTI-184069 (renal cell cancer).
Collapse
Affiliation(s)
- Kenji Hiraizumi
- Oncology Medical Affairs, Ono Pharmaceutical Co., Ltd., 1-8-2 Kyutaromachi, Chuo-ku, Osaka, 541-8564, Japan
| | - Chikara Honda
- PV Data Strategy, Pharmacovigilance Department, Ono Pharmaceutical Co., Ltd., 2-1-5 Dosho-machi, Chuo-ku, Osaka, 541-8526, Japan
| | - Ayu Watanabe
- Safety Management Pharmacovigilance Department, Ono Pharmaceutical Co., Ltd., 2-1-5 Dosho-machi, Chuo-ku, Osaka, 541-8526, Japan
| | - Takafumi Nakao
- Safety Management Pharmacovigilance Department, Ono Pharmaceutical Co., Ltd., 2-1-5 Dosho-machi, Chuo-ku, Osaka, 541-8526, Japan
| | - Shuichi Midorikawa
- Biometrics and Data Sciences, R&D Department, Bristol-Myers Squibb K.K., Otemachi One Tower, 1-2-1 Otemachi, Chiyoda-ku, Tokyo, 100-0004, Japan
| | - Hiromi Abe
- Oncology Medical, Bristol-Myers Squibb K.K., Otemachi One Tower, 1-2-1 Otemachi, Chiyoda-ku, Tokyo, 100-0004, Japan
| | - Nobuki Matsui
- Patient Safety Japan, Bristol-Myers Squibb K.K., Otemachi One Tower, 1-2-1 Otemachi, Chiyoda-ku, Tokyo, 100-0004, Japan
| | - Tsunehisa Yamamoto
- Oncology Medical, Bristol-Myers Squibb K.K., Otemachi One Tower, 1-2-1 Otemachi, Chiyoda-ku, Tokyo, 100-0004, Japan
| | - Takahiko Sakamoto
- Safety Management Pharmacovigilance Department, Ono Pharmaceutical Co., Ltd., 2-1-5 Dosho-machi, Chuo-ku, Osaka, 541-8526, Japan.
| |
Collapse
|
120
|
Huang RJ, Huang YS, An N, Hu JJ, Wu CY, Chen YX, Chen JY, Zhao Q, Xu RH, Yuan SQ, Wang F. Pan-cancer analysis of heterogeneity of tumor mutational burden and genomic mutation under treatment pressure. ESMO Open 2024; 9:103494. [PMID: 38981309 PMCID: PMC11292426 DOI: 10.1016/j.esmoop.2024.103494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/16/2024] [Accepted: 05/07/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND High tumor mutational burden (TMB) is one of the widely researched predictive biomarkers of immune checkpoint inhibitors and has been shown to be closely related with response to immunotherapy in multiple cancer types. However, for patients who have failed conventional therapy and are about to undergo immunotherapy, there is no consensus recommendation on the timing of tumor sampling for TMB analysis, and the effects of different therapies on TMB have not been clarified. This retrospective observational study aimed to investigate the heterogeneity of TMB and genomic mutation under the treatment pressure. PATIENTS AND METHODS We retrospectively collected the available genomic and therapeutic information from 8051 samples across 15 tumor types (>50 samples/tumor) found in 30 published studies and investigated the distribution and heterogeneity of TMB under treatment across diverse cohorts. RESULTS This integrated analysis has shown anticancer treatments increased TMB. Significant effects of treatment on TMB were more frequently observed in tumor types with lower treatment-naïve TMB, including breast, prostate, and pediatric cancers. For different cancer therapies, chemotherapy was prone to be correlated with an increased TMB in most cancer types. Meanwhile, the fraction of the TMB-high category of breast, prostate, and bladder cancers and glioma increased significantly after chemotherapy. Several actionable genes including ERS1 and NF1 in breast cancer, as well as some prognostic markers including TERT in bladder cancer and IDH1 in glioma, were significantly changed in post-chemotherapy tumors compared to treatment-naïve tumors. CONCLUSION Our study reveals the heterogeneity of TMB under treatment across diverse cancer types and provides evidences that chemotherapy was associated with increases in TMB as well as the fraction of TMB-high category, suggesting that resampling tumor tissues for calculating post-chemotherapy TMB could be a better option for predicting the response to immunotherapy, especially for tumors with initially low TMB.
Collapse
Affiliation(s)
- R J Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou
| | - Y S Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou
| | - N An
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou
| | - J J Hu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou
| | - C Y Wu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou
| | - Y X Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou
| | - J Y Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou
| | - Q Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou; Bioinformatic Platform, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou
| | - R H Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou; Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, P. R. China.
| | - S Q Yuan
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou.
| | - F Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou; Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, P. R. China.
| |
Collapse
|
121
|
Yasuda T, Wang YA. Gastric cancer immunosuppressive microenvironment heterogeneity: implications for therapy development. Trends Cancer 2024; 10:627-642. [PMID: 38600020 PMCID: PMC11292672 DOI: 10.1016/j.trecan.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/12/2024]
Abstract
Although immunotherapy has revolutionized solid tumor treatment, durable responses in gastric cancer (GC) remain limited. The heterogeneous tumor microenvironment (TME) facilitates immune evasion, contributing to resistance to conventional and immune therapies. Recent studies have highlighted how specific TME components in GC acquire immune escape capabilities through cancer-specific factors. Understanding the underlying molecular mechanisms and targeting the immunosuppressive TME will enhance immunotherapy efficacy and patient outcomes. This review summarizes recent advances in GC TME research and explores the role of the immune-suppressive system as a context-specific determinant. We also provide insights into potential treatments beyond checkpoint inhibition.
Collapse
Affiliation(s)
- Tadahito Yasuda
- Brown Center for Immunotherapy, Department of Medicine, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Y Alan Wang
- Brown Center for Immunotherapy, Department of Medicine, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
122
|
Sodergren SC, Hurley-Wallace A, Vassiliou V, Alkhaffaf B, Batsaikhan B, Darlington AS, Fleitas-Kanonnikof T, Guren MG, Honda M, Kim YW, Kim S, Krishnamurthy MN, Loh SY, Turhal NS, Zhou J, Dennis K, Krishnatry R, Terashima M, Tsironis G, Yoshikawa T, Terada M. Revisiting the use of the EORTC QLQ-STO22 to assess health-related quality of life of patients with gastric cancer: incorporating updated treatment options and cross-cultural perspectives. Gastric Cancer 2024; 27:722-734. [PMID: 38668819 PMCID: PMC11193686 DOI: 10.1007/s10120-024-01492-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 03/10/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND The EORTC QLQ-STO22 (QLQ-STO22) is a firmly established and validated measure of health-related quality of life (HRQoL) for people with gastric cancer (GC), developed over two decades ago. Since then there have been dramatic changes in treatment options for GC. Also, East Asian patients were not involved in the development of QLQ-STO22, where GC is most prevalent and the QLQ-STO22 is widely used. A review with appropriate updating of the measure was planned. This study aims to capture HRQoL issues associated with new treatments and the perspectives of patients and health care professionals (HCPs) from different cultural backgrounds, including East Asia. METHODS A systematic literature review and open-ended interviews were preformed to identify potential new HRQoL issues relating to GC. This was followed by structured interviews where HCPs and patients reviewed the QLQ-STO22 alongside new issues regarding relevance, importance, and acceptability. RESULTS The review of 267 publications and interviews with 104 patients and 18 HCPs (48 and 9 from East Asia, respectively) generated a list of 58 new issues. Three of these relating to eating small amounts, flatulence, and neuropathy were recommended for inclusion in an updated version of the QLQ-STO22 and covered by five additional questions. CONCLUSIONS This study supports the content validity of the QLQ-STO22, suggesting its continued relevance to patients with GC, including those from East Asia. The updated version with additional questions and linguistic changes will enhance its specificity, but further testing is required.
Collapse
Affiliation(s)
- S C Sodergren
- School of Health Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| | - A Hurley-Wallace
- School of Health Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - V Vassiliou
- Medical Oncology Department, Bank of Cyprus Oncology Centre, Nicosia, Cyprus
| | - B Alkhaffaf
- Department of Oesophago-Gastric & Bariatric Surgery, Northern Care Alliance NHS Foundation Trust, Salford, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, Manchester, UK
| | - B Batsaikhan
- Department of Internal Medicine, Institute of Medical Sciences, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - A S Darlington
- School of Health Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - T Fleitas-Kanonnikof
- Medical Oncology Department, Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - M G Guren
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - M Honda
- General South Tohoku Hospital, Koriyama, Japan
| | - Y W Kim
- Center for Gastric Cancer, National Cancer Center, Goyang-si, Korea
| | - S Kim
- Center for Gastric Cancer, National Cancer Center, Goyang-si, Korea
| | - M N Krishnamurthy
- Department of Clinical Pharmacology, Tata Memorial Centre, Tirupati, India
| | - S Y Loh
- Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - N S Turhal
- Anadolu Medical Center, Çayırova, Turkey
| | - J Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - K Dennis
- Division of Radiation Oncology, The Ottawa Hospital Cancer Centre, Ottawa, Canada
- Department of Radiology, University of Ottawa, Ottawa, Canada
| | - R Krishnatry
- Department of Radiation Oncology, Tata Memorial Centre, Tirupati, India
| | - M Terashima
- Division of Gastric Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - G Tsironis
- Medical Oncology Department, Bank of Cyprus Oncology Centre, Nicosia, Cyprus
| | - T Yoshikawa
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - M Terada
- Department of International Clinical Development/International Trials Management Section, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
123
|
Gao X, Ji K, Jia Y, Shan F, Chen Y, Xu N, Jia Z, Liu T, Yang N, Zhong H, Li C, Guo Z, Fan Q, Lin X, Zhang Y, Ren H, Yang H, Yao Z, Liu W, Wang ZM, Li B, Xia M, Shen L, Li Z, Ji J. Cadonilimab with chemotherapy in HER2-negative gastric or gastroesophageal junction adenocarcinoma: the phase 1b/2 COMPASSION-04 trial. Nat Med 2024; 30:1943-1951. [PMID: 38778212 DOI: 10.1038/s41591-024-03007-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 04/17/2024] [Indexed: 05/25/2024]
Abstract
Treatment with anti-programmed cell death protein 1 (PD-1) therapy and chemotherapy prolongs the survival of patients with unresectable advanced or metastatic gastric or gastroesophageal junction (GEJ) adenocarcinoma. The benefit from anti-PD-1 therapy is enriched in patients with programmed cell death 1 ligand 1 (PD-L1) combined positive score (CPS)-positive or CPS-high tumors compared with patients with PD-L1 CPS-negative or CPS-low tumors. In this phase 1b/2 study, we evaluated the efficacy and safety of cadonilimab, a bispecific antibody targeting PD-1 and cytotoxic T-lymphocyte antigen-4, plus chemotherapy as first-line treatment in patients with human epidermal growth factor receptor 2-negative unresectable advanced or metastatic gastric or GEJ adenocarcinoma. The primary endpoint was the recommended phase 2 dose (RP2D) for phase 1b and the objective response rate for phase 2. Secondary endpoints included disease control rate, duration of response, time to response, progression-free survival, overall survival (OS) and safety. The primary endpoint was met. No dose-limiting toxicities were observed during dose escalation in phase 1b; the recommended phase 2 dose was determined as 6 mg kg-1 every 2 weeks. The objective response rate was 52.1% (95% confidence interval (CI) = 41.6-62.5), consisting of complete and partial responses in 4.3% and 47.9% of patients, respectively. The median duration of response, progression-free survival and OS were 13.73 months (95% CI = 7.79-19.12), 8.18 months (95% CI = 6.67-10.48) and 17.48 months (95% CI = 12.35-26.55), respectively. The median OS in patients with a PD-L1 CPS ≥ 5 was 20.32 months (95% CI = 4.67-not estimable); in patients with a PD-L1 CPS < 1, the median OS reached 17.64 months (95% CI = 11.63-31.70). The most common treatment-related grade 3 or higher adverse events were decreased neutrophil count (19.1%), decreased platelet count (16.0%), anemia (12.8%) and decreased leukocyte count (8.5%). No new safety signal was identified. The current regimen showed promising clinical activity and manageable safety in patients with gastric or GEJ adenocarcinoma regardless of PD-L1 expression. Chinadrugtrials.org.cn registration: CTR20182027.
Collapse
MESH Headings
- Humans
- Stomach Neoplasms/drug therapy
- Stomach Neoplasms/pathology
- Middle Aged
- Male
- Female
- Esophagogastric Junction/pathology
- Adenocarcinoma/drug therapy
- Adenocarcinoma/pathology
- Aged
- Receptor, ErbB-2/metabolism
- Adult
- Esophageal Neoplasms/drug therapy
- Esophageal Neoplasms/pathology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Bispecific/therapeutic use
- Antibodies, Bispecific/adverse effects
- Antibodies, Bispecific/administration & dosage
- B7-H1 Antigen/antagonists & inhibitors
Collapse
Affiliation(s)
- Xiangyu Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ke Ji
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yongning Jia
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Fei Shan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ye Chen
- The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, China
| | - Nong Xu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziyu Jia
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | | | - Nong Yang
- Hunan Cancer Hospital, Changsha, China
| | | | | | | | - Qingxia Fan
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xiaoyan Lin
- Fujian Medical University Union Hospital, Fuzhou, China
| | - Yan Zhang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hui Ren
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hongxia Yang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | | | - Wei Liu
- Akeso Biopharma, Inc, Zhongshan, China
| | | | | | | | - Lin Shen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Ziyu Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Jiafu Ji
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
124
|
Liu P, Wu J, Chen L, Wu Z, Wu Y, Zhang G, Yu B, Zhang B, Wei N, Shi J, Zhang C, Lei L, Yu S, Lai J, Guo Z, Zheng Y, Jing Z, Jiang H, Wang T, Zhou J, Wu Y, Sun C, Shen J, Zhang J, Wu Z. Water-filtered infrared A radiation hyperthermia combined with immunotherapy for advanced gastrointestinal tumours. Cancer Med 2024; 13:e70024. [PMID: 39049187 PMCID: PMC11269209 DOI: 10.1002/cam4.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 06/18/2024] [Accepted: 07/05/2024] [Indexed: 07/27/2024] Open
Abstract
This study pioneered the use of WIRA whole-body infrared hyperthermia combined with ICI therapy to treat GIT and verified the feasibility and safety of HIT. The final results showed a DCR of 55.6%, with a median PFS of 53.5 days, median OS of 134 days, and an irAE incidence of 22.2%. Therefore, we believe that HIT can exert multiple synergistic sensitisation effects, thereby providing clinical benefits to patients with advanced GITs, increasing overall safety, and improving patients' QOL.
Collapse
Affiliation(s)
- Pengyuan Liu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Jing Wu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Liting Chen
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Zhenhai Wu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Yufei Wu
- ACS (International) School of SingaporeSingapore
| | - Ganlu Zhang
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Bingqi Yu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Beibei Zhang
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Nan Wei
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Jinan Shi
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | | | - Lan Lei
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Shuhuan Yu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Jianjun Lai
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Zhen Guo
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Yuli Zheng
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Zhao Jing
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Hao Jiang
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | | | - Jueyi Zhou
- Department of OncologyLishui People's HospitalLishuiChina
| | - Yajun Wu
- TCM Dispensary, Zhejiang HospitalHangzhouChina
| | - Chuan Sun
- Geriatrics Institute of Zhejiang ProvinceDepartment of Geriatrics, Zhejiang HospitalHangzhouChina
| | - Jie Shen
- Department of Medical Oncology, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jian Zhang
- Department of Gastrointestinal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Zhibing Wu
- Department of Oncology, Zhejiang HospitalHangzhouChina
- Department of Radiation Oncology, Affiliated Zhejiang HospitalZhejiang University School of MedicineHangzhouChina
- Cancer CenterZhejiang UniversityHangzhouChina
| |
Collapse
|
125
|
Gao Y, Li H, Qiu L, Yuan H, Fan Q, Niu Z, Xing L, Li M, Yuan D. Efficacy and safety of anti-programmed death-1 antibody-based combination therapy in advanced or metastatic gastric or gastroesophageal junction cancer in Chinese patients: A real-world study. Sci Prog 2024; 107:368504241272703. [PMID: 39166262 PMCID: PMC11339938 DOI: 10.1177/00368504241272703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
PURPOSE Programmed death-1 antibody plus chemotherapy has gained approval for the treatment for (human epidermal growth factor receptor 2 negative locally advanced or metastatic gastric or gastroesophageal junction cancer. This study aims to analyze the efficacy and safety of anti-programmed death-1 antibody combined with chemo- or anti-angiogenesis therapy in Chinese patients with advanced or metastatic gastric or gastroesophageal junction cancer in a real-world setting. METHODS In total, 122 patients treated with anti-programmed death-1 antibody-based combination therapy between April 2019 and December 2021 were encompassed. Clinical outcomes and safety profile were measured and analyzed. RESULTS In the whole cohort, median overall survival was 17.2 months, median progression-free survival was 10.9 months, and median duration of response was 9.4 months. Notably, in the first-line patients, the median overall survival was not reached, median progression-free survival was 14.8 months, objective response rate was 68.4%. In the second-line group, median overall survival, median progression-free survival, median duration of response, and objective response rate were 10.9 months, 5.9 months, 4.5 months, and 41.5%, respectively. Treatment-related adverse events of any grade were observed in 28.2% of the overall cohort, primarily affecting the hematological and liver function. Grade 3 or 4 adverse events were mainly characterized by increased levels of aspartate aminotransferase, alanine aminotransferase, along with decreased lymphocyte and white blood cells, as well as anemia. CONCLUSIONS Patients in our cohort experienced a clinical benefit from anti-programmed death-1 antibody-combined treatment in first-line treatment settings, with acceptable treatment-related adverse events. The benefit of anti-programmed death-1 antibody combined with chemo- or anti-angiogenesis treatment to the second-line patients should be further confirmed by large multi-center randomized, controlled clinical trials.
Collapse
Affiliation(s)
- Yifan Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Oncology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Haoqian Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lei Qiu
- Department of Gastroenterology Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Hongtu Yuan
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Qing Fan
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zuoxing Niu
- Department of Gastroenterology Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Dandan Yuan
- Department of Gastroenterology Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
126
|
Tabuchi M, Kikuchi S, Tazawa H, Okura T, Ogawa T, Mitsui E, Une Y, Kuroda S, Sato H, Noma K, Kagawa S, Ohara T, Ohtsuka J, Ohki R, Urata Y, Fujiwara T. Functional remodeling of intraperitoneal macrophages by oncolytic adenovirus restores anti-tumor immunity for peritoneal metastasis of gastric cancer. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200806. [PMID: 38745748 PMCID: PMC11090911 DOI: 10.1016/j.omton.2024.200806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/29/2024] [Accepted: 04/18/2024] [Indexed: 05/16/2024]
Abstract
Intraperitoneal tumor-associated macrophages (TAMs) are involved in evading anti-tumor immunity and promoting the peritoneal metastasis (PM) of gastric cancer (GC). Oncolytic viruses are known to induce the activation of host anti-tumor immunity in addition to tumor lysis. This study investigated whether a wild-type p53-loading telomerase-specific oncolytic adenovirus (OBP-702) could elicit the remodeling of intraperitoneal macrophages and enhance the efficacy of immune therapy. Increased numbers of CD163 TAMs and few CD8+ lymphocytes were immunohistochemically observed in clinical samples with PM, which suggested that TAMs were associated with the suppression of anti-tumor immunity. OBP-702 induced immunogenic cell death and upregulated PD-L1 expression in human and murine GC cell lines. Intraperitoneal administration of OBP-702 increased recruitment of CD8+ lymphocytes into the PM via the functional remodeling of intraperitoneal macrophages from TAM toward a pro-inflammatory phenotype, resulting in significantly suppressed tumor growth for the in vivo model. Furthermore, the combination of intraperitoneal OBP-702 with anti-programmed cell death-1 antibody enhanced anti-tumor immunity and prolonged the survival of mice bearing PM. Intraperitoneal immunotherapy using OBP-702 restores anti-tumor immunity via the remodeling of intraperitoneal macrophages in addition to direct tumor lysis and cooperates with immune checkpoint inhibitors to suppress PM in GC.
Collapse
Affiliation(s)
- Motoyasu Tabuchi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Satoru Kikuchi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hiroshi Tazawa
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama 700-8558, Japan
| | - Tomohiro Okura
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Toshihiro Ogawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Ema Mitsui
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Yuta Une
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shinji Kuroda
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hiroki Sato
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Kazuhiro Noma
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shunsuke Kagawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Toshiaki Ohara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Junko Ohtsuka
- Laboratory of Fundamental Oncology, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Rieko Ohki
- Laboratory of Fundamental Oncology, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Yasuo Urata
- Oncolys BioPharma, Inc., Tokyo 106-0032, Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
127
|
Yamada E, Iwasaki K, Barroga E, Sakurai T, Enomoto M, Shimoda Y, Mazaki J, Kuwabara H, Hoshino A, Hayashi Y, Ishizaki T, Nagakawa Y. Clinical complete response after trastuzumab deruxtecan 6th-line treatment for postoperative gastric cancer recurrence: a case report. Surg Case Rep 2024; 10:149. [PMID: 38886285 PMCID: PMC11182999 DOI: 10.1186/s40792-024-01954-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Despite the recent developments in the treatment of advanced or recurrent gastric cancer, the median survival time remains shorter than 15 months. Herein, we report a case of postoperative gastric cancer recurrence in which a complete clinical response was achieved with trastuzumab deruxtecan as 6th-line treatment. CASE PRESENTATION A 70-year-old man underwent abdominal contrast-enhanced computed tomography (CT) during follow-up after rectal cancer surgery. The CT revealed an enlarged perigastric lymph node. After further examination, the patient's condition was diagnosed as gastric cancer cT2N1H0P0M0 cStage IIA. The patient underwent distal gastrectomy and D2 lymph node dissection. The resulting pathological diagnosis was pT1bN3aH0P0 pStageIIB, HER2 score 3+. Abdominal contrast-enhanced CT 19 months postoperatively revealed para-aortic lymph node recurrence, thus systemic chemotherapy courses were planned. The primary treatment was a combination of S-1, cisplatin, and trastuzumab administered in 11 courses. However, there was an enlargement of the para-aortic lymph node which was evaluated as progressive disease. Systematic chemotherapy with various regimens was continued until the 5th-line treatment. However, therapeutic benefits were not achieved and lung metastasis was observed. Trastuzumab deruxtecan (TDXD) was initiated as 6th-line treatment. Abdominal contrast-enhanced CT at 4 months after the start of treatment showed marked shrinkage of the enlarged para-aortic lymph node and disappearance of the lung metastasis in the right upper lung lobe, which was evaluated as partial response (PR). The para-aortic lymph node metastasis was evaluated as PR with only a slight accumulation of SUV-Max 2.66 with a shrinking trend by positron emission tomography-computed tomography (PET-CT) performed after 1 year. Tumor markers CEA, CA19-9, and CA125 also improved significantly. PET-CT after 1 year and 4 months showed no lymph node enlargement or accumulation, indicating a complete response (CR). All tumor markers also normalized. The patient has maintained clinical CR without additional treatment to date. CONCLUSIONS We report the apparent first case of postoperative gastric cancer recurrence successfully treated with TDXD, achieving clinical CR with TDXD as a 6th-line treatment.
Collapse
Affiliation(s)
- Erika Yamada
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| | - Kenichi Iwasaki
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan.
| | - Edward Barroga
- Medical English Education Center, Showa University School of Medicine, Tokyo, Japan
| | - Toru Sakurai
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| | - Masaya Enomoto
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| | - Yota Shimoda
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| | - Junichi Mazaki
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| | - Hiroshi Kuwabara
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| | - Akihiro Hoshino
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| | - Yutaka Hayashi
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| | - Tetsuo Ishizaki
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| | - Yuichi Nagakawa
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| |
Collapse
|
128
|
Zeng Z, Zhu Q. Progress and prospects of biomarker-based targeted therapy and immune checkpoint inhibitors in advanced gastric cancer. Front Oncol 2024; 14:1382183. [PMID: 38947886 PMCID: PMC11211377 DOI: 10.3389/fonc.2024.1382183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/24/2024] [Indexed: 07/02/2024] Open
Abstract
Gastric cancer and gastroesophageal junction cancer represent the leading cause of tumor-related death worldwide. Although advances in immunotherapy and molecular targeted therapy have expanded treatment options, they have not significantly altered the prognosis for patients with unresectable or metastatic gastric cancer. A minority of patients, particularly those with PD-L1-positive, HER-2-positive, or MSI-high tumors, may benefit more from immune checkpoint inhibitors and/or HER-2-directed therapies in advanced stages. However, for those lacking specific targets and unique molecular features, conventional chemotherapy remains the only recommended effective and durable regimen. In this review, we summarize the roles of various signaling pathways and further investigate the available targets. Then, the current results of phase II/III clinical trials in advanced gastric cancer, along with the superiorities and limitations of the existing biomarkers, are specifically discussed. Finally, we will offer our insights in precision treatment pattern when encountering the substantial challenges.
Collapse
Affiliation(s)
| | - Qing Zhu
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
129
|
Poniewierska-Baran A, Sobolak K, Niedźwiedzka-Rystwej P, Plewa P, Pawlik A. Immunotherapy Based on Immune Checkpoint Molecules and Immune Checkpoint Inhibitors in Gastric Cancer-Narrative Review. Int J Mol Sci 2024; 25:6471. [PMID: 38928174 PMCID: PMC11203505 DOI: 10.3390/ijms25126471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/31/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Due to its rapid progression to advanced stages and highly metastatic properties, gastric cancer (GC) is one of the most aggressive malignancies and the fourth leading cause of cancer-related deaths worldwide. The metastatic process includes local invasion, metastasis initiation, migration with colonisation at distant sites, and evasion of the immune response. Tumour growth involves the activation of inhibitory signals associated with the immune response, also known as immune checkpoints, including PD-1/PD-L1 (programmed death 1/programmed death ligand 1), CTLA-4 (cytotoxic T cell antigen 4), TIGIT (T cell immunoreceptor with Ig and ITIM domains), and others. Immune checkpoint molecules (ICPMs) are proteins that modulate the innate and adaptive immune responses. While their expression is prominent on immune cells, mainly antigen-presenting cells (APC) and other types of cells, they are also expressed on tumour cells. The engagement of the receptor by the ligand is crucial for inhibiting or stimulating the immune cell, which is an extremely important aspect of cancer immunotherapy. This narrative review explores immunotherapy, focusing on ICPMs and immune checkpoint inhibitors in GC. We also summarise the current clinical trials that are evaluating ICPMs as a target for GC treatment.
Collapse
Affiliation(s)
- Agata Poniewierska-Baran
- Center of Experimental Immunology and Immunobiology of Infectious and Cancer Diseases, University of Szczecin, 71-417 Szczecin, Poland; (A.P.-B.); (P.N.-R.)
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Karolina Sobolak
- Students Research Club of Immunobiology of Infectious and Cancer Diseases “NEUTROPHIL”, University of Szczecin, 71-417 Szczecin, Poland; (K.S.); (P.P.)
| | - Paulina Niedźwiedzka-Rystwej
- Center of Experimental Immunology and Immunobiology of Infectious and Cancer Diseases, University of Szczecin, 71-417 Szczecin, Poland; (A.P.-B.); (P.N.-R.)
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland
| | - Paulina Plewa
- Students Research Club of Immunobiology of Infectious and Cancer Diseases “NEUTROPHIL”, University of Szczecin, 71-417 Szczecin, Poland; (K.S.); (P.P.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
130
|
Ibrahim D, Simó C, Brown EL, Shmuel S, Panikar SS, Benton A, DeWeerd R, Dehdashti F, Park H, Pereira PMR. PD-L1 has a heterogeneous and dynamic expression in gastric cancer with implications for immunoPET. Front Immunol 2024; 15:1405485. [PMID: 38915392 PMCID: PMC11194338 DOI: 10.3389/fimmu.2024.1405485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/21/2024] [Indexed: 06/26/2024] Open
Abstract
Introduction This study aimed to investigate the dynamics of programmed death-ligand 1 (PD-L1) expression, spatial heterogeneity, and binding affinity of FDA-approved anti-PD-L1 antibodies (avelumab and atezolizumab) in gastric cancer. Additionally, we determined how PD-L1 glycosylation impacts antibody accumulation in gastric cancer cells. Methods Dynamic PD-L1 expression was examined in NCIN87 gastric cancer cells. Comparative binding studies of avelumab and atezolizumab were conducted in gastric cancer models, both in vitro and in vivo. Antibody uptake in tumors was visualized through positron emission tomography (PET) imaging. PD-L1 glycosylation status was determined via Western blot analyses before and after PNGase F treatment. Results Consistent findings revealed time-dependent PD-L1 induction in NCIN87 gastric cancer cells and spatial heterogeneity in tumors, as shown by PET imaging and immunofluorescence. Avelumab displayed superior binding affinity to NCIN87 cells compared to atezolizumab, confirmed by in vivo PET imaging and ex vivo biodistribution analyses. Notably, PD-L1 glycosylation at approximately 50 kDa was observed, with PNGase F treatment inducing a shift to 35 kDa in molecular weight. Tissue samples from patient-derived xenografts (PDXs) validated the presence of both glycosylated and deglycosylated PD-L1 (degPD-L1) forms in gastric cancer. Immunofluorescence microscopy and binding assays demonstrated enhanced avelumab binding post-deglycosylation. Discussion This study provides an understanding of dynamic and spatially heterogeneous PD-L1 expression in gastric cancer. Anti-PD-L1 immunoPET was able to visualize gastric tumors, and PD-L1 glycosylation has significant implications for antibody recognition. These insights contribute to demonstrating the complexities of PD-L1 in gastric cancer, holding relevance for refining PD-L1 imaging-based approaches.
Collapse
Affiliation(s)
- Dina Ibrahim
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Cristina Simó
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Emma L. Brown
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Shayla Shmuel
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Sandeep Surendra Panikar
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Alex Benton
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
- Cancer Biology Graduate Program, Washington University School of Medicine, St. Louis, MO, United States
| | - Rachel DeWeerd
- Cancer Biology Graduate Program, Washington University School of Medicine, St. Louis, MO, United States
| | - Farrokh Dehdashti
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Haeseong Park
- Gastrointestinal Cancer Center, Center for Cancer Therapeutic Innovation, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Patrícia M. R. Pereira
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
131
|
Sekihara K, Himuro H, Toda S, Saito N, Hirayama R, Suganuma N, Sasada T, Hoshino D. Recent Trends and Potential of Radiotherapy in the Treatment of Anaplastic Thyroid Cancer. Biomedicines 2024; 12:1286. [PMID: 38927493 PMCID: PMC11201408 DOI: 10.3390/biomedicines12061286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Anaplastic thyroid cancer (ATC) is a rare but highly aggressive malignancy characterized by advanced disease at diagnosis and a poor prognosis. Despite multimodal therapeutic approaches that include surgery, radiotherapy, and chemotherapy, an optimal treatment strategy remains elusive. Current developments in targeted therapies and immunotherapy offer promising avenues for improved outcomes, particularly for BRAF-mutant patients. However, challenges remain regarding overcoming drug resistance and developing effective treatments for BRAF-wild-type tumors. This comprehensive review examines the clinical and biological features of ATC, outlines the current standards of care, and discusses recent developments with a focus on the evolving role of radiotherapy. Moreover, it emphasizes the necessity of a multidisciplinary approach and highlights the urgent need for further research to better understand ATC pathogenesis and identify new therapeutic targets. Collaborative efforts, including large-scale clinical trials, are essential for translating these findings into improved patient outcomes.
Collapse
Affiliation(s)
- Kazumasa Sekihara
- Cancer Biology Division, Kanagawa Cancer Center Research Institute, Yokohama 2418515, Japan; (K.S.); (S.T.); (N.S.)
- Biospecimen Center, Kanagawa Cancer Center, Yokohama 2418515, Japan
| | - Hidetomo Himuro
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 2418515, Japan; (H.H.); (T.S.)
- Department of Radiation Oncology, Kanagawa Cancer Center, Yokohama 2418515, Japan
| | - Soji Toda
- Cancer Biology Division, Kanagawa Cancer Center Research Institute, Yokohama 2418515, Japan; (K.S.); (S.T.); (N.S.)
- Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, Yokohama 2320024, Japan
| | - Nao Saito
- Cancer Biology Division, Kanagawa Cancer Center Research Institute, Yokohama 2418515, Japan; (K.S.); (S.T.); (N.S.)
- Biospecimen Center, Kanagawa Cancer Center, Yokohama 2418515, Japan
| | - Ryoichi Hirayama
- Department of Charged Particle Therapy Research, QST Hospital, National Institutes for Quantum Science and Technology, Chiba 2638555, Japan;
| | - Nobuyasu Suganuma
- Department of Surgery, Yokohama City University, Yokohama 2360004, Japan;
| | - Tetsuro Sasada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 2418515, Japan; (H.H.); (T.S.)
| | - Daisuke Hoshino
- Cancer Biology Division, Kanagawa Cancer Center Research Institute, Yokohama 2418515, Japan; (K.S.); (S.T.); (N.S.)
- Biospecimen Center, Kanagawa Cancer Center, Yokohama 2418515, Japan
| |
Collapse
|
132
|
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. [PMID: 38832979 PMCID: PMC11150231 DOI: 10.1007/s00262-024-03721-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/02/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND The beneficial effects of first-line programmed death-1 (PD-1) inhibitors plus chemotherapy in patients with low programmed death-ligand 1 (PD-L1)-expressing advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma are controversial. METHODS We conducted a retrospective analysis of patients with G/GEJ adenocarcinoma who had undergone first-line treatment with PD-1 inhibitors plus chemotherapy between October 2017 and May 2022. The primary outcomes were objective response rate (ORR) and progression-free survival (PFS). SPSS software V27.0 was used for data analysis. RESULTS Of 345 enrolled patients, 290 had measurable lesions. The overall ORR was 59.3%. PD-L1 status was available in 171 patients, and 67.8% of them were considered as low PD-L1 expression level (combined positive score (CPS) < 5). Patients with PD-L1 CPS < 5 showed a lower response rate (51.1% vs 70.8%, P = 0.024) and a worse PFS (P = 0.009) compared to those with PD-L1 CPS ≥ 5. In the PD-L1 low-expression cohort, patients with non-diffuse type, GEJ cancer, synchronous metastasis, distant lymph node metastasis, liver metastasis, non-peritoneal metastasis, and HER2 positive were significantly associated with higher response rates to PD-1 inhibitors plus chemotherapy (P < 0.05). The presence of peritoneal metastasis (P = 0.028) and diffuse type (P = 0.046) were identified as independent predictors of poor PFS in multivariate analysis of the PD-L1 CPS < 5 subgroup. When evaluated for correlation with overall survival (OS) in the PD-L1 low-expression subgroup, peritoneal metastasis was found to be the only independent prognostic factor of an increased risk of death (hazard ratio: 2.31, 95% CI 1.09-4.90; P = 0.029). CONCLUSIONS PD-L1 CPS ≥ 5 is significantly associated with improved response and extended PFS in G/GEJ cancer patients treated with a combination of PD-1 inhibitors and chemotherapy. Specific subgroups within the low PD-L1-expressing population, such as those with non-diffuse-type tumors and without peritoneal metastases, may also benefit from immunotherapy combined with chemotherapy.
Collapse
Affiliation(s)
- Yu-Ting Sun
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Shi-Xun Lu
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
| | - Ming-Yu Lai
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
| | - Xia Yang
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
| | - Wen-Long Guan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
| | - Li-Qiong Yang
- Department of Basic Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
| | - Yu-Hong Li
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
| | - Feng-Hua Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
| | - Da-Jun Yang
- Department of Basic Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China.
| | - Miao-Zhen Qiu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
133
|
Kadono T, Iwasa S, Hirose T, Hirano H, Okita N, Shoji H, Takashima A, Kato K. Impact of immune checkpoint inhibitors on survival outcomes in advanced gastric cancer in Japan: A real-world analysis. Cancer Med 2024; 13:e7401. [PMID: 38899745 PMCID: PMC11187802 DOI: 10.1002/cam4.7401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/14/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Nivolumab was approved for the treatment of advanced gastric cancer in 2017 in Japan. The aim of this study was to assess the impact of nivolumab in a real-world clinical setting. METHODS This single-institutional retrospective study included patients with advanced gastric or esophagogastric junction adenocarcinoma and a history of first-line chemotherapy with platinum-based doublet or triplet regimens between 2010 and 2020. To assess the impact of nivolumab on survival, the patients were divided based on the year of nivolumab approval into a pre-2017 (2010-2016) group and a post-2017 (2017-2020) group. RESULTS From a total of 1918 patients, 1093 were excluded. There were 533 patients in the pre-2017 group and 292 in the post-2017 group. Immune checkpoint inhibitors were used significantly more often in the post-2017 group than in the pre-2017 group (8.6% vs. 47.9%). Median overall survival was significantly longer in the post-2017 group (16.9 vs. 13.9 months; hazard ratio [HR] 0.75, 95% confidence interval [CI] 0.63-0.90; p < 0.01). The proportion of patients transitioning to third-line treatment was higher in the post-2017 group than in the pre-2017 group (56.3% vs. 43.8%, p < 0.01). Median survival outcomes following progression on second-line treatment were significantly longer in the post-2017 group (4.3 vs. 3.2 months; HR 0.70, 95% CI 0.57-0.86; p < 0.01). CONCLUSION The proportion of patients transitioning to third-line treatment and survival outcomes following progression on second-line treatment have improved since the approval of nivolumab. This drug might help to prolong overall survival in real-world practice.
Collapse
Affiliation(s)
- Toru Kadono
- Department of Gastrointestinal Medical OncologyNational Cancer Center HospitalTokyoJapan
- Cancer Chemotherapy CenterOsaka Medical and Pharmaceutical UniversityTakatsukiOsakaJapan
| | - Satoru Iwasa
- Department of Gastrointestinal Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Toshiharu Hirose
- Department of Gastrointestinal Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Hidekazu Hirano
- Department of Gastrointestinal Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Natsuko Okita
- Department of Gastrointestinal Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Hirokazu Shoji
- Department of Gastrointestinal Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Atsuo Takashima
- Department of Gastrointestinal Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Ken Kato
- Department of Gastrointestinal Medical OncologyNational Cancer Center HospitalTokyoJapan
| |
Collapse
|
134
|
Jazieh K, Yoon H, Zhu M. Advances in Immunotherapy in Esophagogastric Cancer. Hematol Oncol Clin North Am 2024; 38:599-616. [PMID: 38493074 DOI: 10.1016/j.hoc.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2024]
Abstract
Immune checkpoint inhibitors are rapidly transforming the care of patients with esophagogastric cancer. Particularly, anti-PD-1 therapy has demonstrated promising efficacy in metastatic and resectable disease. In this review, the authors discuss landmark clinical trials, highlight challenges and opportunities in this field, and propose potential directions for future work.
Collapse
Affiliation(s)
- Khalid Jazieh
- Division of Medical Oncology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA.
| | - Harry Yoon
- Division of Medical Oncology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Mojun Zhu
- Division of Medical Oncology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| |
Collapse
|
135
|
Ikejiri F, Yokomizo K, Tamura K. Successful treatment with nivolumab in a patient with gastric cancer with severe liver failure resulting from multiple liver metastases: A case report. Oncol Lett 2024; 27:271. [PMID: 38686354 PMCID: PMC11057026 DOI: 10.3892/ol.2024.14404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/05/2024] [Indexed: 05/02/2024] Open
Abstract
Gastric cancer (GC) is a globally prevalent and deadly malignancy often diagnosed at advanced stages, which can be accompanied by liver metastases. Conventional chemotherapy is contraindicated in patients with severe liver failure because several chemotherapeutic agents are metabolized by the liver. The present study reports on the successful use of nivolumab in a patient with advanced GC and severe liver failure owing to multiple liver metastases. A 57-year-old man was admitted to Shimane Prefectural Central Hospital (Izumo, Japan) with a 2-week history of appetite loss and jaundice. An upper gastrointestinal endoscopy revealed advanced GC (type IV). Computed tomography examination confirmed wall thickening of the gastric pylorus and the presence of multiple liver metastases. A gastric mucosal biopsy confirmed the diagnosis of HER2-positive gastric adenocarcinoma. S-1 + cisplatin chemotherapy was initiated but had to be halted due to the rapid deterioration in liver function, ultimately leading to acute liver failure. The patient was discharged from the hospital under palliative care. The patient was referred to Shimane University Hospital (Izumo, Japan) for a second consultation. Upon admission, the patient presented with severe liver failure, a Child-Pugh score of 10 (Class C), elevated total bilirubin levels of 13.9 mg/dl (normal range: <1.8 mg/dl) and elevated CEA and CA19-9. Nivolumab treatment was initiated, and notably, there was a substantial reduction in bilirubin levels, an improvement in liver function after a single cycle and a partial response observed in imaging studies. Despite the initial poor prognosis, the patient achieved long-term survival, ultimately succumbing to the illness 2 years and 6 months following the initiation of treatment. The present case underscores the potential of immune checkpoint inhibitors, such as nivolumab, in the treatment of patients with cancer and severe liver failure. It also challenges the conventional constraints of chemotherapy, offering a promising direction for future research in this area.
Collapse
Affiliation(s)
- Fumiyoshi Ikejiri
- Department of Medical Oncology, Shimane University Hospital Innovative Cancer Center, Izumo, Shimane 693-8501, Japan
- Department of Internal Medicine, Okuizumo Town Hospital, Okuizumo, Shimane 699-1511, Japan
| | - Kanako Yokomizo
- Department of Medical Oncology, Shimane University Hospital Innovative Cancer Center, Izumo, Shimane 693-8501, Japan
- Clinical Training Center, Tsuyama Chuo Hospital, Tsuyama, Okayama, 708-0841, Japan
| | - Kenji Tamura
- Department of Medical Oncology, Shimane University Hospital Innovative Cancer Center, Izumo, Shimane 693-8501, Japan
| |
Collapse
|
136
|
Alcindor T. Immunotherapy in Gastric Cancer-Choosing Methods or Results. JAMA Oncol 2024; 10:704-705. [PMID: 38573642 DOI: 10.1001/jamaoncol.2023.7262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Affiliation(s)
- Thierry Alcindor
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
137
|
Tsuji K, Miyajima S, Kito Y. Nivolumab Rechallenge After Prior Nivolumab Therapy in Advanced Gastric Cancer: A Single-Center Case Series and Literature Review. J Gastrointest Cancer 2024; 55:956-960. [PMID: 38165606 DOI: 10.1007/s12029-023-01011-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2023] [Indexed: 01/04/2024]
Abstract
BACKGROUND AND AIMS Pivotal phase III trials indicated that the anti-PD-1 inhibitor nivolumab prolongs overall survival in patients with advanced gastric cancer. Nivolumab is currently used in the first- or later-line treatment of patients with advanced gastric cancer in Japan. The efficacy of immune check inhibitor rechallenge after progression has been reported in other cancers. Therefore, this study investigated the clinical outcome of nivolumab rechallenge in patients with advanced gastric cancer who received nivolumab in a previous systemic line. METHODS We retrospectively reviewed the medical records of six patients with advanced or recurrent gastric cancer who received nivolumab rechallenge. RESULTS During initial nivolumab therapy, three patients experienced partial responses, and one patient achieved stable disease. The reasons for discontinuing initial nivolumab therapy were progressive disease in five patients and immune-related adverse events in one patient. The median interval duration of treatment for patients receiving both nivolumab regimens was 13.7 (range: 5.1-17.8) months. During nivolumab rechallenge, no patients achieved partial responses, whereas two patients had stable disease. Median progression-free survival was 2.5 (95% confidence interval [CI] = 1.6-not available [NA]) months, and median overall survival was 7.4 (95% CI = 2.3-NA) months. Although one patient had discontinued prior nivolumab therapy because of immune-related adverse events, there were no immune-related adverse events associated with nivolumab rechallenge. CONCLUSIONS The benefit of nivolumab rechallenge in patients with advanced gastric cancer was limited. Rechallenge with the same immune check inhibitor might be ineffective in patients with advanced gastric cancer.
Collapse
Affiliation(s)
- Kunihiro Tsuji
- Department of Medical Oncology, Ishikawa Prefectural Central Hospital, 2-1 Kuratukihigashi, 920-8530, Kanazawa, Ishikawa, Japan.
| | - Saori Miyajima
- Department of Medical Oncology, Ishikawa Prefectural Central Hospital, 2-1 Kuratukihigashi, 920-8530, Kanazawa, Ishikawa, Japan
| | - Yosuke Kito
- Department of Medical Oncology, Ishikawa Prefectural Central Hospital, 2-1 Kuratukihigashi, 920-8530, Kanazawa, Ishikawa, Japan
| |
Collapse
|
138
|
Zhang W, Wang X, Dong J, Wang K, Jiang W, Fan C, Liu H, Fan L, Zhao L, Li G. Single-cell analysis uncovers high-proliferative tumour cell subtypes and their interactions in the microenvironment of gastric cancer. J Cell Mol Med 2024; 28:e18373. [PMID: 38894657 PMCID: PMC11187953 DOI: 10.1111/jcmm.18373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/07/2024] [Accepted: 04/18/2024] [Indexed: 06/21/2024] Open
Abstract
Gastric cancer (GC) remains a prominent malignancy that poses a significant threat to human well-being worldwide. Despite advancements in chemotherapy and immunotherapy, which have effectively augmented patient survival rates, the mortality rate associated with GC remains distressingly high. This can be attributed to the elevated proliferation and invasive nature exhibited by GC. Our current understanding of the drivers behind GC cell proliferation remains limited. Hence, in order to reveal the molecular biological mechanism behind the swift advancement of GC, we employed single-cell RNA-sequencing (scRNA-seq) to characterize the tumour microenvironment in this study. The scRNA-seq data of 27 patients were acquired from the Gene Expression Omnibus database. Differential gene analysis, Gene Ontology, Kyoto Encyclopedia of Genes and Genomes and Gene Set Enrichment Analysis were employed to investigate 38 samples. The copy number variation level exhibited by GC cells was determined using InferCNV. The CytoTRACE, Monocle and Slingshot analysis were used to discern the cellular stemness and developmental trajectory of GC cells. The CellChat package was utilized for the analysis of intercellular communication crosstalk. Moreover, the findings of the data analysis were validated through cellular functional tests conducted on the AGS cell line and SGC-7901 cell line. Finally, this study constructed a risk scoring model to evaluate the differences of different risk scores in clinical characteristics, immune infiltration, immune checkpoints, functional enrichment, tumour mutation burden and drug sensitivity. Within the microenvironment of GC, we identified the presence of 8 cell subsets, encompassing NK_T cells, B_Plasma cells, epithelial cells, myeloid cells, endothelial cells, mast cells, fibroblasts, pericytes. By delving deeper into the characterization of GC cells, we identified 6 specific tumour cell subtypes: C0 PSCA+ tumour cells, C1 CLDN7+ tumour cells, C2 UBE2C+ tumour cells, C3 MUC6+ tumour cells, C4 CHGA+ tumour cells and C5 MUC2+ tumour cells. Notably, the C2 UBE2C+ tumour cells demonstrated a close association with cell mitosis and the cell cycle, exhibiting robust proliferative capabilities. Our findings were fortified through enrichment analysis, pseudotime analysis and cell communication analysis. Meanwhile, knockdown of the transcription factor CREB3, which is highly active in UBE2C+ tumour cells, significantly impedes the proliferation, migration and invasion of GC cells. And the prognostic score model constructed with CREB3-related genes showcased commendable clinical predictive capacity, thus providing valuable guidance for patients' prognosis and clinical treatment decisions. We have identified a highly proliferative cellular subgroup C2 UBE2C+ tumour cells in GC for the first time. The employment of a risk score model, which is based on genes associated with UBE2C expression, exhibits remarkable proficiency in predicting the prognosis of GC patients. In our investigation, we observed that the knockdown of the transcription factor CREB3 led to a marked reduction in cellular proliferation, migration and invasion in GC cell line models. Implementing a stratified treatment approach guided by this model represents a judicious and promising methodology.
Collapse
Affiliation(s)
- Wenjia Zhang
- Department of Respiratory Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Xiaojing Wang
- Department of Rheumatology and Immunology, Tongren Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jiaxing Dong
- Department of Gastrointestinal Surgery, Shanghai East Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Kai Wang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of MedicineTongji UniversityShanghaiChina
| | - Wanju Jiang
- Department of Gastrointestinal Surgery, Shanghai East Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Chenchen Fan
- Department of Respiratory Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Haitao Liu
- Department of Respiratory Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Lihong Fan
- Department of Respiratory Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Lei Zhao
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of MedicineTongji UniversityShanghaiChina
| | - Guoshu Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of MedicineTongji UniversityShanghaiChina
| |
Collapse
|
139
|
Sun J, Li X, Wang Q, Chen P, Zhao L, Gao Y. Proteomic profiling and biomarker discovery for predicting the response to PD-1 inhibitor immunotherapy in gastric cancer patients. Front Pharmacol 2024; 15:1349459. [PMID: 38881867 PMCID: PMC11176556 DOI: 10.3389/fphar.2024.1349459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 05/08/2024] [Indexed: 06/18/2024] Open
Abstract
Background: Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment; however, a significant proportion of gastric cancer (GC) patients do not respond to this therapy. Consequently, there is an urgent need to elucidate the mechanisms underlying resistance to ICIs and identify robust biomarkers capable of predicting the response to ICIs at treatment initiation. Methods: In this study, we collected GC tissues from 28 patients prior to the administration of anti-programmed death 1 (PD-1) immunotherapy and conducted protein quantification using high-resolution mass spectrometry (MS). Subsequently, we analyzed differences in protein expression, pathways, and the tumor microenvironment (TME) between responders and non-responders. Furthermore, we explored the potential of these differences as predictive indicators. Finally, using machine learning algorithms, we screened for biomarkers and constructed a predictive model. Results: Our proteomics-based analysis revealed that low activity in the complement and coagulation cascades pathway (CCCP) and a high abundance of activated CD8 T cells are positive signals corresponding to ICIs. By using machine learning, we successfully identified a set of 10 protein biomarkers, and the constructed model demonstrated excellent performance in predicting the response in an independent validation set (N = 14; area under the curve [AUC] = 0.959). Conclusion: In summary, our proteomic analyses unveiled unique potential biomarkers for predicting the response to PD-1 inhibitor immunotherapy in GC patients, which may provide the impetus for precision immunotherapy.
Collapse
Affiliation(s)
- Jiangang Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaojing Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qian Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Longfei Zhao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yongshun Gao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
140
|
Khan B, Qahwaji RM, Alfaifi MS, Mobashir M. Nivolumab and Ipilimumab Acting as Tormentors of Advanced Tumors by Unleashing Immune Cells and Associated Collateral Damage. Pharmaceutics 2024; 16:732. [PMID: 38931856 PMCID: PMC11207028 DOI: 10.3390/pharmaceutics16060732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 06/28/2024] Open
Abstract
Combining immune checkpoint inhibitors, specifically nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4), holds substantial promise in revolutionizing cancer treatment. This review explores the transformative impact of these combinations, emphasizing their potential for enhancing therapeutic outcomes across various cancers. Immune checkpoint proteins, such as PD1 and CTLA4, play a pivotal role in modulating immune responses. Blocking these checkpoints unleashes anticancer activity, and the synergy observed when combining multiple checkpoint inhibitors underscores their potential for enhanced efficacy. Nivolumab and ipilimumab harness the host's immune system to target cancer cells, presenting a powerful approach to prevent tumor development. Despite their efficacy, immune checkpoint inhibitors are accompanied by a distinct set of adverse effects, particularly immune-related adverse effects affecting various organs. Understanding these challenges is crucial for optimizing treatment strategies and ensuring patient well-being. Ongoing clinical trials are actively exploring the combination of checkpoint inhibitory therapies, aiming to decipher their synergistic effects and efficacy against diverse cancer types. This review discusses the mechanisms, adverse effects, and various clinical trials involving nivolumab and ipilimumab across different cancers, emphasizing their transformative impact on cancer treatment.
Collapse
Affiliation(s)
- Bushra Khan
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Rowaid M. Qahwaji
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22233, Saudi Arabia;
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mashael S. Alfaifi
- Department of Epidemiology, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Mohammad Mobashir
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, Solnavägen 9, 171 65 Solna, Sweden
| |
Collapse
|
141
|
Matsuda K, Shimazu K, Shinozaki H, Fukuda K, Yoshida T, Taguchi D, Nomura K, Shibata H. Recent trends in bone metastasis treatments: A historical comparison using the new Katagiri score system. World J Clin Cases 2024; 12:2499-2505. [PMID: 38817228 PMCID: PMC11135455 DOI: 10.12998/wjcc.v12.i15.2499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 04/06/2024] [Accepted: 04/15/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND Bone metastasis has various negative impacts. Activities of daily living (ADL) and quality of life (QOL) can be significantly decreased, survival may be impacted, and medical expenses may increase. It is estimated that at least 5% cancer patients might be suffering from bone metastases. In 2016, we published the Comprehensive Guidelines for the Diagnosis and Treatment of Bone Metastasis. Since then, the therapeutic outcomes for patients have gradually improved. As life expectancy is a major determinant of surgical intervention, the strategy should be modified if the prolongation of survival is to be achieved. AIM To monitor how bone metastasis treatment has changed before and after launch of our guidelines for bone metastasis. METHODS For advanced cancer patients with bone metastasis who visited the Department of Clinical Oncology at Akita University hospital between 2012 and 2023, parameters including the site and number of bone metastases, laboratory data, and survival time, were extracted from electronic medical records and the Katagiri score was calculated. The association with survival was determined for each factor. RESULTS Data from 136 patients were obtained. The 1-year survival rate for the poor prognosis group with a higher Katagiri score was 20.0% in this study, which was 6% and an apparent improvement from 2014 when the scoring system was developed. Other factors significantly affecting survival included five or more bone metastases than less (P = 0.0080), and treatment with chemotherapy (P < 0.001), bone modifying agents (P = 0.0175) and immune checkpoint inhibitors (P = 0.0128). In recent years, advances in various treatment methods have extended the survival period for patients with advanced cancer. It is necessary not only to simply extend survival time, but also to maintain ADL and improve QOL. CONCLUSION Various therapeutic interventions including surgical approach for bone metastasis, which is a disorder of locomotor organs, are increasingly required. Guidelines and scoring system for prognosis need to be revised promptly.
Collapse
Affiliation(s)
- Kenji Matsuda
- Department of Clinical Oncology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Kazuhiro Shimazu
- Department of Clinical Oncology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Hanae Shinozaki
- Department of Clinical Oncology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Koji Fukuda
- Department of Clinical Oncology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Taichi Yoshida
- Department of Clinical Oncology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Daiki Taguchi
- Department of Clinical Oncology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Kyoko Nomura
- Department of Environmental Health Science and Public Health, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Hiroyuki Shibata
- Department of Clinical Oncology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
- Department of Comprehensive Cancer Control, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| |
Collapse
|
142
|
Meng X, Bai X, Ke A, Li K, Lei Y, Ding S, Dai D. Long Non-Coding RNAs in Drug Resistance of Gastric Cancer: Complex Mechanisms and Potential Clinical Applications. Biomolecules 2024; 14:608. [PMID: 38927012 PMCID: PMC11201466 DOI: 10.3390/biom14060608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 05/11/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Gastric cancer (GC) ranks as the third most prevalent malignancy and a leading cause of cancer-related mortality worldwide. However, the majority of patients with GC are diagnosed at an advanced stage, highlighting the urgent need for effective perioperative and postoperative chemotherapy to prevent relapse and metastasis. The current treatment strategies have limited overall efficacy because of intrinsic or acquired drug resistance. Recent evidence suggests that dysregulated long non-coding RNAs (lncRNAs) play a significant role in mediating drug resistance in GC. Therefore, there is an imperative to explore novel molecular mechanisms underlying drug resistance in order to overcome this challenging issue. With advancements in deep transcriptome sequencing technology, lncRNAs-once considered transcriptional noise-have garnered widespread attention as potential regulators of carcinogenesis, including tumor cell proliferation, metastasis, and sensitivity to chemo- or radiotherapy through multiple regulatory mechanisms. In light of these findings, we aim to review the mechanisms by which lncRNAs contribute to drug therapy resistance in GC with the goal of providing new insights and breakthroughs toward overcoming this formidable obstacle.
Collapse
Affiliation(s)
- Xiangyu Meng
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; (X.M.); (X.B.); (K.L.); (Y.L.); (S.D.)
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang 110042, China
| | - Xiao Bai
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; (X.M.); (X.B.); (K.L.); (Y.L.); (S.D.)
| | - Angting Ke
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; (X.M.); (X.B.); (K.L.); (Y.L.); (S.D.)
| | - Kaiqiang Li
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; (X.M.); (X.B.); (K.L.); (Y.L.); (S.D.)
| | - Yun Lei
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; (X.M.); (X.B.); (K.L.); (Y.L.); (S.D.)
| | - Siqi Ding
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; (X.M.); (X.B.); (K.L.); (Y.L.); (S.D.)
| | - Dongqiu Dai
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; (X.M.); (X.B.); (K.L.); (Y.L.); (S.D.)
- Cancer Center, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| |
Collapse
|
143
|
Kadono T, Yamamoto S, Kato K. Development of perioperative immune checkpoint inhibitor therapy for locally advanced esophageal squamous cell carcinoma. Future Oncol 2024; 20:2097-2107. [PMID: 38861290 PMCID: PMC11497952 DOI: 10.1080/14796694.2024.2345043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 04/16/2024] [Indexed: 06/12/2024] Open
Abstract
The standard preoperative treatment for resectable locally advanced esophageal squamous cell carcinoma (ESCC) is chemoradiotherapy in western countries (based on the CROSS trial) and triplet chemotherapy in Japan (based on the JCOG1109 trial). Postoperative nivolumab has recently been shown to improve disease-free survival in resectable locally advanced esophageal cancer after preoperative chemoradiotherapy in patients who had residual pathological disease, based on the CheckMate 577 trial. Furthermore, preoperative immune checkpoint inhibitor-containing treatments have also been developed. The JCOG1804E trial is presently evaluating the safety and efficacy of preoperative nivolumab-containing chemotherapy for resectable locally advanced ESCC. This review discusses the treatment of resectable locally advanced ESCC and future perspectives on perioperative immune checkpoint inhibitor-containing treatments.
Collapse
Affiliation(s)
- Toru Kadono
- Department of Head & Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
- Cancer Chemotherapy Center, Osaka Medical & Pharmaceutical University, Takatsuki, Osaka, 569-8686, Japan
| | - Shun Yamamoto
- Department of Head & Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Ken Kato
- Department of Head & Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| |
Collapse
|
144
|
Wang Z, Li X, Hu J, Guo X, Gao B, Zhu B. Bibliometric and visual analysis of esophagogastric junction cancer research from 2002 to 2021. Medicine (Baltimore) 2024; 103:e38100. [PMID: 38758908 PMCID: PMC11098202 DOI: 10.1097/md.0000000000038100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/11/2024] [Indexed: 05/19/2024] Open
Abstract
Numerous studies related to esophagogastric junction cancer (EGC) have been published, and bibliometric analysis of these publications may be able to identify research hotspots and frontiers of EGC. Studies published on EGC between 2002 and 2021 were retrieved from the Web of Science Core Collection. The collaboration network of countries/regions, institutions, authors, co-citation network of journals, co-occurrence network, and overlay visualization of keywords were analyzed using the VOSviewer software. Cluster and timeline analyses of references were performed using the CiteSpace software. A total of 5109 English articles were published across 691 journals by authors affiliated with 4727 institutions from 81 countries/regions. The annual number of publications related to EGC research has exhibited an increasing trend. The United States, China, and Japan emerged as the top 3 prolific countries/regions. Institutions in the United States, Japan, and South Korea exhibited significant collaboration with one another. Diseases of the Esophagus was the most prolific journal, and Annals of Surgical Oncology, World Journal of Gastroenterology, and Gastric Cancer had also published more than 100 studies. Jaffer A Ajani was the most productive author while David Cunningham ranked the first in terms of total citations and average citations per article. Barrett's esophagus, gastroesophageal reflux disease, Helicobacter pylori, and obesity were common topics in earlier research, and recent years had seen a shift towards the topics of immunotherapy, targeted therapy, and neoadjuvant chemotherapy. In conclusion, growing attention is paid to EGC research, especially in terms of immunotherapy, targeted therapy, and neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Zhuoyin Wang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinming Li
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jili Hu
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xu Guo
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Bulang Gao
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Bin Zhu
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
145
|
Wang J, Lin J, Wang R, Tong T, Zhao Y. Immunotherapy combined with apatinib in the treatment of advanced or metastatic gastric/gastroesophageal tumors: a systematic review and meta-analysis. BMC Cancer 2024; 24:603. [PMID: 38760737 PMCID: PMC11102247 DOI: 10.1186/s12885-024-12340-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/06/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Immunotherapy or apatinib alone has been used as third-line adjuvant therapy for advanced or metastatic gastric/gastroesophageal junction (G/GEJ) tumors, but the efficacy of combining them with each other for the treatment of patients with advanced or metastatic G/GEJ is unknown; therefore, we further evaluated the efficacy and safety of immunotherapy combined with apatinib in patients with advanced or metastatic G/GEJ. METHODS The main search was conducted on published databases: Embase, Cochrane library, PubMed.The search was conducted from the establishment of the database to December 2023.Clinical trials with patients with advanced or metastatic G/GEJ and immunotherapy combined with apatinib as the study variable were collected. Review Manager 5.4 software as well as stata 15.0 software were used for meta-analysis. RESULTS A total of 651 patients from 19 articles were included in this meta-analysis. In the included studies, immunotherapy combined with apatinib had a complete response (CR) of 0.03 (95% CI: 0.00 -0.06), partial response (PR) of 0.34 (95% CI: 0.19-0.49), stable disease (SD) of 0.43 (95% CI: 0.32-0.55), objective response rate (ORR) was 0.36 (95% CI: 0.23-0.48), disease control rate (DCR) was 0.80 (95% CI: 0.74-0.86), and median progression-free survival (PFS) was 4.29 (95% CI: 4.05-4.52), median Overall survival (OS) was 8.79 (95% CI: 7.92-9.66), and the incidence of grade ≥ 3 TRAEs was 0.34 (95% CI: 0:19-0.49). PR, ORR, DCR, median PFS and median OS were significantly higher in the immunotherapy and apatinib combination chemotherapy group (IAC) than in the immunotherapy combination apatinib group (IA). And the difference was not significant in the incidence of SD and grade ≥ 3 TRAEs. CONCLUSION This meta-analysis shows that immunotherapy combined with apatinib is safe and effective in the treatment of advanced or metastatic G/GEJ, where IAC can be a recommended adjuvant treatment option for patients with advanced or metastatic G/GEJ. However, more large multicenter randomized studies are urgently needed to reveal the long-term outcomes of immunotherapy combined with apatinib treatment.
Collapse
Affiliation(s)
- Jincheng Wang
- Department of Thoracic Surgery, the Second Hospital of Jilin University, Changchun City, China
| | - Jie Lin
- Department of Hepatobiliary and Pancreatic Surgery, the Second Hospital of Jilin University, Changchun City, 130000, Jilin, China
| | - Ruimin Wang
- Department of Operating Room, The Second Hospital of Jilin University, Changchun City, 130041, Jilin, China
| | - Ti Tong
- Department of Thoracic Surgery, the Second Hospital of Jilin University, Changchun City, China
| | - Yinghao Zhao
- Department of Thoracic Surgery, the Second Hospital of Jilin University, Changchun City, China.
| |
Collapse
|
146
|
Lin F, Chen Y, Huang B, Ruan S, Lin J, Chen Z, Huang C, Zhao B. Application of immune checkpoint inhibitors for resectable gastric/gastroesophageal cancer. Front Pharmacol 2024; 15:1391562. [PMID: 38783944 PMCID: PMC11111861 DOI: 10.3389/fphar.2024.1391562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024] Open
Abstract
Gastric/gastroesophageal junction (G/GEJ) cancer represents a significant global health challenge. Radical surgery remains the cornerstone of treatment for resectable G/GEJ cancer. Supported by robust evidence from multiple clinical studies, therapeutic approaches, including adjuvant chemotherapy or chemoradiation, and perioperative chemotherapy, are generally recommended to reduce the risk of recurrence and enhance long-term survival outcomes post-surgery. In recent years, immune checkpoint inhibitors (ICIs) have altered the landscape of systemic treatment for advanced or metastatic G/GEJ cancer, becoming the standard first-line therapy for specific patients. Consequently, exploring the efficacy of ICIs in the adjuvant or neoadjuvant setting for resectable G/GEJ cancer is worthwhile. This review summarizes the current advances in the application of ICIs for resectable G/GEJ cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Chunyu Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Baiwei Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
147
|
Zhao P, Zhao T, Yu L, Ma W, Liu W, Zhang C. The risk of endocrine immune-related adverse events induced by PD-1 inhibitors in cancer patients: a systematic review and meta-analysis. Front Oncol 2024; 14:1381250. [PMID: 38756658 PMCID: PMC11096456 DOI: 10.3389/fonc.2024.1381250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 04/17/2024] [Indexed: 05/18/2024] Open
Abstract
Objective Endocrinopathies are the most common immune-related adverse events (irAEs) observed during therapy with PD-1 inhibitors. In this study, we conducted a comprehensive systematic review and meta-analysis to evaluate the risk of immune-related endocrinopathies in patients treated with PD-1 inhibitors. Methods We performed a systematic search in the PubMed, Embase, and Cochrane Library databases to retrieve all randomized controlled trials (RCTs) involving PD-1 inhibitors, spanning from their inception to November 24, 2023. The comparative analysis encompassed patients undergoing chemotherapy, targeted therapy, or receiving placebo as control treatments. This study protocol has been registered with PROSPERO (CRD42023488303). Results A total of 48 clinical trials comprising 24,514 patients were included. Compared with control groups, patients treated with PD-1 inhibitors showed an increased risk of immune-related adverse events, including hypothyroidism, hyperthyroidism, hypophysitis, thyroiditis, diabetes mellitus, and adrenal insufficiency. Pembrolizumab was associated with an increased risk of all aforementioned endocrinopathies (hypothyroidism: RR=4.76, 95%CI: 3.55-6.39; hyperthyroidism: RR=9.69, 95%CI: 6.95-13.52; hypophysitis: RR=5.47, 95%CI: 2.73-10.97; thyroiditis: RR=5.95, 95%CI: 3.02-11.72; diabetes mellitus: RR=3.60, 95%CI: 1.65-7.88; adrenal insufficiency: RR=4.80, 95%CI: 2.60-8.88). Nivolumab was associated with an increased risk of hypothyroidism (RR=7.67, 95%CI: 5.00-11.75) and hyperthyroidism (RR=9.22, 95%CI: 4.71-18.04). Tislelizumab and sintilimab were associated with an increased risk of hypothyroidism (RR=19.07, 95%CI: 5.46-66.69 for tislelizumab and RR=18.36, 95%CI: 3.58-94.21 for sintilimab). For different tumor types, both hypothyroidism and hyperthyroidism were at high risks. Besides, patients with non-small cell lung cancer were at a higher risk of thyroiditis and adrenal insufficiency. Patients with melanoma were at a higher risk of hypophysitis and diabetes mellitus. Both low- and high-dose group increased risks of hypothyroidism and hyperthyroidism. Conclusion Risk of endocrine irAEs may vary in different PD-1 inhibitors and different tumor types. Increased awareness and understanding of the risk features of endocrine irAEs associated with PD-1 inhibitors is critical for clinicians. Systematic review registration crd.york.ac.uk/prospero, identifier PROSPERO (CRD42023488303).
Collapse
Affiliation(s)
- Pengfei Zhao
- Department of Clinical Pharmacy, Weifang People's Hospital, Weifang, China
| | - Ting Zhao
- Department of Clinical Pharmacy, Weifang People's Hospital, Weifang, China
| | - Lihong Yu
- Department of Clinical Pharmacy, Weifang People's Hospital, Weifang, China
| | - Wenming Ma
- Department of Clinical Pharmacy, Weifang People's Hospital, Weifang, China
| | - Wenyu Liu
- Department of Pharmacy, Weifang People's Hospital, Weifang, China
| | - Chenning Zhang
- Department of Rehabilitation Medicine & Department of Pharmacy, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| |
Collapse
|
148
|
Zhao Y, Li D, Zhuang J, Li Z, Xia Q, Li Z, Yu J, Wang J, Zhang Y, Li K, Xu S, Li S, Ma P, Cao Y, Liu C, Xu C, Liu Z, Wei J, Zhang C, Qiao L, Gao X, Hou Z, Liu C, Zheng R, Wang D, Liu Y. Comprehensive multi-omics analysis of resectable locally advanced gastric cancer: Assessing response to neoadjuvant camrelizumab and chemotherapy in a single-center, open-label, single-arm phase II trial. Clin Transl Med 2024; 14:e1674. [PMID: 38685486 PMCID: PMC11058238 DOI: 10.1002/ctm2.1674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/07/2024] [Accepted: 04/11/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND The current standard of care for locally advanced gastric cancer (GC) involves neoadjuvant chemotherapy followed by radical surgery. Recently, neoadjuvant treatment for this condition has involved the exploration of immunotherapy plus chemotherapy as a potential approach. However, the efficacy remains uncertain. METHODS A single-arm, phase 2 study was conducted to evaluate the efficacy and tolerability of neoadjuvant camrelizumab combined with mFOLFOX6 and identify potential biomarkers of response through multi-omics analysis in patients with resectable locally advanced GC. The primary endpoint was the pathological complete response (pCR) rate. Secondary endpoints included the R0 rate, near pCR rate, progression-free survival (PFS), disease-free survival (DFS), and overall survival (OS). Multi-omics analysis was assessed by whole-exome sequencing, transcriptome sequencing, and multiplex immunofluorescence (mIF) using biopsies pre- and post-neoadjuvant therapy. RESULTS This study involved 60 patients, of which 55 underwent gastrectomy. Among these, five (9.1%) attained a pathological complete response (pCR), and 11 (20.0%) reached near pCR. No unexpected treatment-emergent adverse events or perioperative mortality were observed, and the regimen presented a manageable safety profile. Molecular changes identified through multi-omics analysis correlated with treatment response, highlighting associations between HER2-positive and CTNNB1 mutations with treatment sensitivity and a favourable prognosis. This finding was further supported by immune cell infiltration analysis and mIF. Expression data uncovered a risk model with four genes (RALYL, SCGN, CCKBR, NTS) linked to poor response. Additionally, post-treatment infiltration of CD8+ T lymphocytes positively correlates with pathological response. CONCLUSION The findings suggest the combination of PD-1-inhibitor and mFOLFOX6 showed efficacy and acceptable toxicity for locally advanced GC. Extended follow-up is required to determine the duration of the response. This study lays essential groundwork for developing precise neoadjuvant regimens.
Collapse
Affiliation(s)
- Yuzhou Zhao
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Danyang Li
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Jing Zhuang
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Zhimeng Li
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Qingxin Xia
- Department of PathologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Zhi Li
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Juan Yu
- Department of Endoscopy CenterThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Jinbang Wang
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Yong Zhang
- Department of ImmunotherapyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Ke Li
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Shuning Xu
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Sen Li
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Pengfei Ma
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Yanghui Cao
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Chenyu Liu
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Chunmiao Xu
- Department of RadiologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Zhentian Liu
- Department of translational medicineGeneplus‐Beijing InstituteBeijingChina
| | - Jinwang Wei
- GenomiCare Biotechnology LA Co., Ltd.ShanghaiChina
| | - Chengjuan Zhang
- Department of PathologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Lei Qiao
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Xuan Gao
- Department of translational medicineGeneplus‐Beijing InstituteBeijingChina
| | - Zhiguo Hou
- Jiangsu Hengrui Pharmaceuticals Co., Ltd.ShanghaiChina
| | - Chenxuan Liu
- Jiangsu Hengrui Pharmaceuticals Co., Ltd.ShanghaiChina
| | | | - Du Wang
- Jiangsu Hengrui Pharmaceuticals Co., Ltd.ShanghaiChina
| | - Ying Liu
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| |
Collapse
|
149
|
Takeno A, Motoori M, Kishi K, Omori T, Hirao M, Masuzawa T, Fujitani K, Yamamato K, Kurokawa Y, Doki Y. Prognostic factors of conversion surgery for stage IV gastric cancer: A multi-institutional retrospective analysis. Ann Gastroenterol Surg 2024; 8:431-442. [PMID: 38707233 PMCID: PMC11066490 DOI: 10.1002/ags3.12778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/06/2024] [Accepted: 01/13/2024] [Indexed: 05/07/2024] Open
Abstract
Background Conversion surgery (CS) is a highly anticipated strategy for stage IV advanced gastric cancer (AGC) with a good response to chemotherapy. However, prognostic factors limiting R0 resection remain unclear. In this multi-institutional study, we investigated the clinical outcomes of CS for stage IV AGC and the prognostic factors of CS-limiting R0 resection and analyzed them according to metastatic patterns. Methods Clinical data on 210 patients who underwent CS for stage IV AGC at six institutions between 2007 and 2017 were retrospectively retrieved. The patient background, preoperative treatment, operative outcomes, and survival times were recorded. Prognostic factors for overall and recurrence-free survival were investigated using univariate and multivariate analyses for patients who underwent R0 resection. Results R0 resection was achieved in 146 (70%) patients. The median survival time was 32 months, and the 3-year survival rate was 45%. Patients who achieved R0 resection had significantly longer survival than those with R1/2 resection (median survival time: 41.5 months vs. 20.7 months). Multivariate analysis identified pathological N positivity for overall and relapse-free survival and pathological T4 for relapse-free survival as significant independent poor prognostic factors of R0 resected patients. There was no significant difference in survival among the peritoneum, liver, and lymph node groups regarding the initial metastatic sites. Conclusions CS with R0 resection for patients with stage IV AGC can lead to longer survival. Patients with pathological T4 and pathological N positivity were eligible for intensive adjuvant therapy after CS with R0 resection.
Collapse
Affiliation(s)
- Atsushi Takeno
- Department of Surgery, National Hospital OrganizationOsaka National HospitalOsakaJapan
| | | | - Kentaro Kishi
- Department of SurgeryOsaka Police HospitalOsakaJapan
| | - Takeshi Omori
- Department of Gastroenterological SurgeryOsaka International Cancer InstituteOsakaJapan
| | - Motohiro Hirao
- Department of Surgery, National Hospital OrganizationOsaka National HospitalOsakaJapan
| | - Toru Masuzawa
- Department of SurgeryKansai Rosai HospitalAmagasakiHyogoJapan
| | | | - Kazuyoshi Yamamato
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversitySuitaOsakaJapan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversitySuitaOsakaJapan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversitySuitaOsakaJapan
| |
Collapse
|
150
|
Ito Y, Kanda M, Sasahara M, Tanaka C, Shimizu D, Umeda S, Inokawa Y, Hattori N, Hayashi M, Nakayama G, Kodera Y. Killer cell lectin-like receptor G2 facilitates aggressive phenotypes of gastric cancer cells via dual activation of the ERK1/2 and JAK/STAT pathways. Gastric Cancer 2024; 27:506-518. [PMID: 38386237 DOI: 10.1007/s10120-024-01480-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 02/06/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Advanced gastric cancer (GC) has a poor prognosis. This study aimed to identify novel GC-related genes as potential therapeutic targets. METHODS Killer cell lectin-like receptor G2 (KLRG2) was identified as a candidate gene by transcriptome analysis of metastatic GC tissues. Small interfering RNA-mediated KLRG2 knockdown in human GC cell lines was used to investigate KLRG2 involvement in signaling pathways and functional behaviors in vitro and in vivo. Clinicopathological data were analyzed in patients stratified according to tumor KLRG2 mRNA expression. RESULTS KLRG2 knockdown in GC cells decreased cell proliferation, migration, and invasion; caused cell cycle arrest in G2/M phase; induced apoptosis via caspase activation; suppressed JAK/STAT and MAPK-ERK1/2 pathway activities; and upregulated p53 and p38 MAPK activities. In mouse xenograft models of peritoneal metastasis, the number and weight of disseminated GC nodules were decreased by KLRG2 knockdown. High tumor levels of KLRG2 mRNA were significantly associated with lower 5-year overall survival (OS) and relapse-free survival (RFS) rates in patients with Stage I-III GC (5-year OS rate: 64.4% vs. 80.0%, P = 0.009; 5-year RFS rate: 62.8% vs. 78.1%, P = 0.030). CONCLUSIONS KLRG2 knockdown attenuated the malignant phenotypes of GC cells via downregulation of JAK/STAT and MAPK-ERK1/2 pathway activity and upregulation of p38 MAPK and p53. Targeted suppression of KLRG2 may serve as a new treatment approach for GC.
Collapse
Affiliation(s)
- Yuki Ito
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan.
| | - Masahiro Sasahara
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Chie Tanaka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Dai Shimizu
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Shinichi Umeda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Yoshikuni Inokawa
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Norifumi Hattori
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Masamichi Hayashi
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Goro Nakayama
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| |
Collapse
|