1
|
Yamazawa S, Fukasawa-Hokazono M, Takase A, Kondo A, Matsubara J, Shinozaki-Ushiku A, Seto Y, Ushiku T. Immune evasion strategies in AFP-producing gastric carcinoma: characterized by HLA-G expression and HLA class I deficiency. Virchows Arch 2025:10.1007/s00428-025-04108-3. [PMID: 40278871 DOI: 10.1007/s00428-025-04108-3] [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: 02/20/2025] [Revised: 03/28/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025]
Abstract
Alpha-fetoprotein-producing gastric carcinoma (AFPGC) is an aggressive subtype of gastric cancer characterized by a primitive cellular phenotype and poor prognosis. The tumor immunology of AFPGC remains largely unexplored. Given its embryonic-like properties, AFPGC is hypothesized to employ distinct immune evasion strategies, with the oncofetal protein human leukocyte antigen (HLA)-G-a key mediator of maternal-fetal immune tolerance-likely playing a pivotal role. To test this, we assessed the expression of HLA-G, along with other key immune evasion markers, including HLA class I (HLA-I) deficiency and PD-L1 expression, in 39 cases of AFPGC, and compared them with those of 44 Epstein-Barr virus (EBV)-positive, 57 microsatellite instability (MSI), 54 intestinal-type, and 45 diffuse-type gastric carcinomas. HLA-G expression was significantly higher in AFPGCs (71%) than in other subtypes (7-28%; P < 0.001). HLA-I deficiency (≥ 1% of tumor cells) was most prevalent in AFPGC (69%), followed by MSI tumors (56%), with lower rates in other subtypes (22-29%). PD-L1 positivity (combined positive score ≥ 5) was observed in 41% of AFPGCs, lower than in EBV-positive (77%) and MSI tumors (44%), but higher than in intestinal-type (13%) and diffuse-type (9%) carcinomas. Furthermore, CD8-positive T-cell infiltration was found to be lowest in AFPGC compared to the other subtypes. These findings suggest that AFPGC employs multiple immune evasion mechanisms, notably through increased HLA-G expression and HLA-I deficiency, likely linked to its primitive cellular phenotype and reactivation of immunogenic oncofetal antigens. Such immune evasion features may underlie the aggressiveness of AFPGC and present promising targets for immunotherapeutic interventions.
Collapse
Affiliation(s)
- Sho Yamazawa
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Akiko Takase
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsushi Kondo
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Joji Matsubara
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Aya Shinozaki-Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Division of Integrative Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
2
|
Wang L, Wu Q, Zhang ZW, Zhang H, Jin H, Zhou XL, Liu JY, Li D, Liu Y, Fan ZS. Colony-stimulating factor 3 and its receptor promote leukocyte immunoglobulin-like receptor B2 expression and ligands in gastric cancer. World J Gastrointest Oncol 2025; 17:97858. [PMID: 39958563 PMCID: PMC11756009 DOI: 10.4251/wjgo.v17.i2.97858] [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: 06/11/2024] [Revised: 10/29/2024] [Accepted: 11/08/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Colony-stimulating factor 3 (CSF3) and its receptor (CSF3R) are known to promote gastric cancer (GC) growth and metastasis. However, their effects on the immune microenvironment remain unclear. Our analysis indicated a potential link between CSF3R expression and the immunosuppressive receptor leukocyte immunoglobulin-like receptor B2 (LILRB2) in GC. We hypothesized that CSF3/CSF3R may regulate LILRB2 and its ligands, angiopoietin-like protein 2 (ANGPTL2) and human leukocyte antigen-G (HLA-G), contributing to immunosuppression. AIM To investigate the relationship between CSF3/CSF3R and LILRB2, as well as its ligands ANGPTL2 and HLA-G, in GC. METHODS Transcriptome sequencing data from The Cancer Genome Atlas were analyzed, stratifying patients by CSF3R expression. Differentially expressed genes and immune checkpoints were evaluated. Immunohistochemistry (IHC) was performed on GC tissues. Correlation analyses of CSF3R, LILRB2, ANGPTL2, and HLA-G were conducted using The Cancer Genome Atlas data and IHC results. GC cells were treated with CSF3, and expression levels of LILRB2, ANGPTL2, and HLA-G were measured by quantitative reverse transcriptase-polymerase chain reaction and western blotting. RESULTS Among 122 upregulated genes in high CSF3R expression groups, LILRB2 showed the most significant increase. IHC results indicated high expression of LILRB2 (63.0%), ANGPTL2 (56.5%), and HLA-G (73.9%) in GC tissues. Strong positive correlations existed between CSF3R and LILRB2, ANGPTL2, and HLA-G mRNA levels (P < 0.001). IHC confirmed positive correlations between CSF3R and LILRB2 (P < 0.001), and HLA-G (P = 0.010), but not ANGPTL2 (P > 0.05). CSF3 increased LILRB2, ANGPTL2, and HLA-G expression in GC cells. Heterogeneous nuclear ribonucleoprotein H1 modulation significantly altered their expression, impacting CSF3's regulatory effects. CONCLUSION The CSF3/CSF3R pathway may contribute to immunosuppression in GC by upregulating LILRB2 and its ligands, with heterogeneous nuclear ribonucleoprotein H1 playing a regulatory role.
Collapse
Affiliation(s)
- Long Wang
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Qi Wu
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Zong-Wen Zhang
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Hui Zhang
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Hui Jin
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Xin-Liang Zhou
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Jia-Yin Liu
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Dan Li
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Yan Liu
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Zhi-Song Fan
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| |
Collapse
|
3
|
Wu JW, Zhou CF, Han ZX, Zhang H, Yan J, Chen J, Wang CB, Qin ZQ, Mao Y, Tang XY, Zhu LJ, Wei XW, Cui DH, Yang XL, Shi M, Zhao LQ, Jiang JL, Zhu WY, Wang HM, Wang C, Zhu LJ, Zhang J. Anlotinib plus chemotherapy as a first-line treatment for gastrointestinal cancer patients with unresectable liver metastases: a multicohort, multicenter, exploratory trial. Signal Transduct Target Ther 2024; 9:344. [PMID: 39648217 PMCID: PMC11625826 DOI: 10.1038/s41392-024-02051-4] [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: 07/31/2024] [Revised: 10/26/2024] [Accepted: 11/04/2024] [Indexed: 12/10/2024] Open
Abstract
This multicohort phase II trial (ALTER-G-001; NCT05262335) aimed to assess the efficacy of first-line anlotinib plus chemotherapy for gastrointestinal (GI) cancer patients with unresectable liver metastases. Eligible patients with colorectal cancer (Cohort A) or noncolorectal and nonesophageal GI cancer (Cohort C) received six cycles of anlotinib plus standard chemotherapeutic regimens followed by anlotinib plus metronomic capecitabine as a maintenance therapy. Liver metastasectomy can be performed when liver metastases are converted to resectable lesions. The primary outcome was the investigator-confirmed objective response rate (ORR) in the intention-to-treat population. Among the 47 patients in Cohort A, the ORR was 40.4% (95% CI 26.4-55.7), including 1 with a complete response (CR) and 18 who achieved a partial response (PR). The median progression-free survival (PFS) was 8.7 months (95% CI 7.3-NE), and the median overall survival (OS) was not reached. In Cohort C, 14 of 44 patients achieved a PR, with an ORR of 31.8% (95% CI 18.6-47.6). The PFS and OS were 5.8 months (95% CI 4.8-6.5) and 11.4 months (95% CI 5.8-19.3), respectively. The liver metastasectomy rate in patients with liver-limited disease was 22.7% (5/22) in Cohort A and 6.7% (2/30) in Cohort C. For pancreatic cancer patients, the ORR of the efficacy-evaluable population was 36.0% (9/25), and those with liver-limited metastasis had better survival. Moreover, no new safety concerns emerged. In conclusion, an anlotinib-based first-line regimen demonstrated promising antitumor activity among GI cancer patients with unresectable liver metastases and led to liver metastasectomy in selected patients.
Collapse
Affiliation(s)
- Jun-Wei Wu
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen-Fei Zhou
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng-Xiang Han
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Huan Zhang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Yan
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Jun Chen
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Chun-Bin Wang
- Department of Oncology, The Third People's Hospital of Yancheng, Yancheng, China
| | - Zhi-Quan Qin
- Department of Medical Oncology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou, China
| | - Yong Mao
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xin-Yu Tang
- Department of Oncology, Wuxi Branch of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Wuxi, China
| | - Liang-Jun Zhu
- Department of Medical Oncology, Jiangsu Cancer Hospital, Nanjing, China
| | - Xiao-Wei Wei
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Dong-Hai Cui
- Department of Internal Medicine, Anyang Tumor Hospital, Anyang, China
| | - Xiu-Li Yang
- Department of Oncology, First Affiliated Hospital of Nanyang Medical College, Nanyang, China
| | - Min Shi
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-Qin Zhao
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin-Ling Jiang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-You Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hong-Mei Wang
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chun Wang
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Ling-Jun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
4
|
Da X, Juan Z, Zhijun H, Zhongchuan L. Case report: Significant response of alpha-fetoprotein-producing gastric cancer from combined chemotherapy and immunotherapy. Front Immunol 2024; 15:1448875. [PMID: 39530092 PMCID: PMC11551777 DOI: 10.3389/fimmu.2024.1448875] [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: 06/14/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Background Alpha-fetoprotein-producing gastric cancer (AFPGC) represents a particularly aggressive subtype of gastric carcinoma characterized by elevated rates of vascular invasion, lymphatic dissemination, hepatic metastasis, and an unfavorable clinical outcome. Treatment strategies for AFPGC have historically lacked specificity. Herein, a case is presented involving AFPGC in which the patient exhibited a notable response to combined anti-PD-1 antibody immunotherapy and SOX chemotherapy, potentially achieving a cure. This report marks the first application of this regimen in neoadjuvant therapy for AFP gastric cancer, followed by radical resection and postoperative adjuvant therapy. Case summary A 62-year-old male patient presented with persistent upper abdominal distension and discomfort lasting over 2 months. Initial investigations revealed markedly elevated serum alpha-fetoprotein (AFP) levels, and subsequent pathological examination confirmed the diagnosis of AFPGC via gastroscopy. Due to the patient's condition, surgical resection was initially deemed unfeasible. Therefore, a chemo-immunotherapy regimen consisting of SOX chemotherapy and the PD-1 inhibitor tislelizumab was administered for 3 cycles. Following this, successful laparoscopic radical gastrectomy was performed. The treatment protocol was continued with an additional 3 cycles postoperatively. At the time of this case report, the patient maintained a good quality of life with no evidence of disease recurrence or adverse events. Conclusion The present report highlights a case of AFPGC where significant therapeutic success was achieved through a combined regimen of chemotherapy and immunotherapy, both before and after surgery. The use of anti-PD-1 antibody (tislelizumab) in combination with SOX regimen (S-1 and oxaliplatin) demonstrated effective treatment of AFPGC, potentially offering a curative approach. This approach represents a promising targeted therapy option for patients with AFPGC.
Collapse
Affiliation(s)
- Xue Da
- Department of General Surgery, Yantai Yuhuangding Hospital, Shandong, China
| | - Zhang Juan
- Department of Pharmacy, Yantai Yuhuangding Hospital, Shandong, China
| | - Hu Zhijun
- Department of General Surgery, Yantai Yuhuangding Hospital, Shandong, China
| | - Lyu Zhongchuan
- Department of General Surgery, Yantai Yuhuangding Hospital, Shandong, China
| |
Collapse
|
5
|
Hu X, Ouyang W, Chen H, Liu Z, Lai Z, Yao H. Claudin-9 (CLDN9) promotes gastric cancer progression by enhancing the glycolysis pathway and facilitating PD-L1 lactylation to suppress CD8+ T cell anti-tumor immunity. CANCER PATHOGENESIS AND THERAPY 2024. [DOI: 10.1016/j.cpt.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/31/2024]
|
6
|
Hao W, Liu W, Chang R, Yang M, Xin K, Liu J, Wang Y, Ren M, Xie J, Yang Y. Safety and clinical efficacy of immune checkpoint inhibitors in advanced gastric cancer in the real world. J Cancer Res Clin Oncol 2024; 150:180. [PMID: 38587528 PMCID: PMC11001672 DOI: 10.1007/s00432-024-05703-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 03/14/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND To evaluate the clinical efficacy and safety of immune checkpoint inhibitors in patients with advanced gastric cancer in the real world. METHODS The retrospective analysis was conducted on the clinical records of 402 patients with advanced gastric cancer who were admitted to the Nanjing Drum Tower Hospital between December 2017 and April 2022 and who had received immunotherapy. Observation target: drug use, treatment, adverse reaction type and grade, objective response rate (ORR), disease control rate (DCR), progression free survival (PFS), and overall survival (OS). RESULTS By retrospectively analyzing the data of patients with advanced gastric cancer treated with ICIs previously admitted to our medical center, we found some clinical characteristic factors associated with the occurrence of irAEs as well as the efficacy and prognosis: the presence or absence of hypertension, whether or not to receive targeted therapies can predict the occurrence of immune-related adverse events (irAEs), and the more the presence of irAEs, the better the prognosis. These can help clinicians in clinical drug selection. CONCLUSIONS The results of this paper show that the occurrence of irAEs is associated with patients' OS. irAEs occurrence can prolong patients' OS. irAEs occurrence may serve as a surrogate marker for ICIs.
Collapse
Affiliation(s)
- Wen Hao
- Department of Oncology, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenjing Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ruimin Chang
- Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Mi Yang
- Department of Oncology, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Kai Xin
- Department of Oncology, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jingxin Liu
- Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yibing Wang
- Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Meijin Ren
- Department of Oncology, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiaqi Xie
- Department of Oncology, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Yang Yang
- Department of Oncology, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
7
|
Liu J, Yuan Q, Guo H, Guan H, Hong Z, Shang D. Deciphering drug resistance in gastric cancer: Potential mechanisms and future perspectives. Biomed Pharmacother 2024; 173:116310. [PMID: 38394851 DOI: 10.1016/j.biopha.2024.116310] [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: 11/24/2023] [Revised: 02/07/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Gastric cancer (GC) is a malignant tumor that originates from the epithelium of the gastric mucosa. The latest global cancer statistics show that GC ranks fifth in incidence and fourth in mortality among all cancers, posing a serious threat to public health. While early-stage GC is primarily treated through surgery, chemotherapy is the frontline option for advanced cases. Currently, commonly used chemotherapy regimens include FOLFOX (oxaliplatin + leucovorin + 5-fluorouracil) and XELOX (oxaliplatin + capecitabine). However, with the widespread use of chemotherapy, an increasing number of cases of drug resistance have emerged. This article primarily explores the potential mechanisms of chemotherapy resistance in GC patients from five perspectives: cell death, tumor microenvironment, non-coding RNA, epigenetics, and epithelial-mesenchymal transition. Additionally, it proposes feasibility strategies to overcome drug resistance from four angles: cancer stem cells, tumor microenvironment, natural products, and combined therapy. The hope is that this article will provide guidance for researchers in the field and bring hope to more GC patients.
Collapse
Affiliation(s)
- Jiahua Liu
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qihang Yuan
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui Guo
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hewen Guan
- First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Zhijun Hong
- First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Dong Shang
- First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
8
|
Jung M, Lee JB, Kim HS, Kwon WS, Kim HO, Kim S, Park M, Kim W, Choi KY, Oh T, Kang CY, Chung HC, Rha SY. First-in-Human Phase 1 Study of a B Cell- and Monocyte-Based Immunotherapeutic Vaccine against HER2-Positive Advanced Gastric Cancer. Cancer Res Treat 2024; 56:208-218. [PMID: 37402409 PMCID: PMC10789970 DOI: 10.4143/crt.2022.1328] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 06/26/2023] [Indexed: 07/06/2023] Open
Abstract
PURPOSE BVAC-B is an autologous B cell- and monocyte-based immunotherapeutic vaccine that contains cells transfected with a recombinant human epidermal growth factor receptor 2 (HER2) gene and loaded with the natural killer T cell ligand alpha-galactosylceramide. Here, we report the first BVAC-B study in patients with HER2-positive advanced gastric cancer. MATERIALS AND METHODS Patients with advanced gastric cancer refractory to standard treatment with HER2+ immunohistochemistry ≥ 1 were eligible for treatment. Patients were administered low (2.5×107 cells/dose), medium (5.0×107 cells/dose), or high dose (1.0×108 cells/dose) of BVAC-B intravenously four times every 4 weeks. Primary endpoints included safety and maximum tolerated BVAC-B dose. Secondary endpoints included preliminary clinical efficacy and BVAC-B-induced immune responses. RESULTS Eight patients were treated with BVAC-B at low (n=1), medium (n=1), and high doses (n=6). No dose-limiting toxicity was observed, while treatment-related adverse events (TRAEs) were observed in patients treated with medium and high doses. The most common TRAEs were grade 1 (n=2) and grade 2 (n=2) fever. Out of the six patients treated with high-dose BVAC-B, three had stable disease with no response. Interferon gamma, tumor necrosis factor-α, and interleukin-6 increased after BVAC-B treatment in all patients with medium and high dose, and HER2-specific antibody was detected in some patients. CONCLUSION BVAC-B monotherapy had a safe toxicity profile with limited clinical activity; however, it activated immune cells in heavily pretreated patients with HER2-positive gastric cancer. Earlier treatment with BVAC-B and combination therapy is warranted for evaluation of clinical efficacy.
Collapse
Affiliation(s)
- Minkyu Jung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University of College of Medicine, Seoul,
Korea
- Song-dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul,
Korea
| | - Jii Bum Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University of College of Medicine, Seoul,
Korea
| | - Hyo Song Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University of College of Medicine, Seoul,
Korea
- Song-dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul,
Korea
| | - Woo Sun Kwon
- Song-dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul,
Korea
| | - Hyun Ok Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul,
Korea
| | - Sinyoung Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul,
Korea
| | | | | | | | | | - Chang-Yuil Kang
- Cellid, Inc., Seoul,
Korea
- Laboratory of Immunology, College of Pharmacy, Seoul National University, Seoul,
Korea
| | - Hyun Cheol Chung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University of College of Medicine, Seoul,
Korea
- Song-dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul,
Korea
| | - Sun Young Rha
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University of College of Medicine, Seoul,
Korea
- Song-dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul,
Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul,
Korea
| |
Collapse
|
9
|
Huang W, Xiong W, Tang L, Chen C, Yuan Q, Zhang C, Zhou K, Sun Z, Zhang T, Han Z, Feng H, Liang X, Zhong Y, Deng H, Yu L, Xu Y, Wang W, Shen L, Li G, Jiang Y. Non-invasive CT imaging biomarker to predict immunotherapy response in gastric cancer: a multicenter study. J Immunother Cancer 2023; 11:e007807. [PMID: 38179695 PMCID: PMC10668251 DOI: 10.1136/jitc-2023-007807] [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] [Accepted: 10/24/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Despite remarkable benefits have been provided by immune checkpoint inhibitors in gastric cancer (GC), predictions of treatment response and prognosis remain unsatisfactory, making identifying biomarkers desirable. The aim of this study was to develop and validate a CT imaging biomarker to predict the immunotherapy response in patients with GC and investigate the associated immune infiltration patterns. METHODS This retrospective study included 294 GC patients who received anti-PD-1/PD-L1 immunotherapy from three independent medical centers between January 2017 and April 2022. A radiomics score (RS) was developed from the intratumoral and peritumoral features on pretreatment CT images to predict immunotherapy-related progression-free survival (irPFS). The performance of the RS was evaluated by the area under the time-dependent receiver operating characteristic curve (AUC). Multivariable Cox regression analysis was performed to construct predictive nomogram of irPFS. The C-index was used to determine the performance of the nomogram. Bulk RNA sequencing of tumors from 42 patients in The Cancer Genome Atlas was used to investigate the RS-associated immune infiltration patterns. RESULTS Overall, 89 of 294 patients (median age, 57 years (IQR 48-66 years); 171 males) had an objective response to immunotherapy. The RS included 13 CT features that yielded AUCs of 12-month irPFS of 0.787, 0.810 and 0.785 in the training, internal validation, and external validation 1 cohorts, respectively, and an AUC of 24-month irPFS of 0.805 in the external validation 2 cohort. Patients with low RS had longer irPFS in each cohort (p<0.05). Multivariable Cox regression analyses showed RS is an independent prognostic factor of irPFS. The nomogram that integrated the RS and clinical characteristics showed improved performance in predicting irPFS, with C-index of 0.687-0.778 in the training and validation cohorts. The CT imaging biomarker was associated with M1 macrophage infiltration. CONCLUSION The findings of this prognostic study suggest that the non-invasive CT imaging biomarker can effectively predict immunotherapy outcomes in patients with GC and is associated with innate immune signaling, which can serve as a potential tool for individual treatment decisions.
Collapse
Affiliation(s)
- Weicai Huang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Wenjun Xiong
- Department of Gastrointestinal Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lei Tang
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Chuanli Chen
- Department of Medical Imaging Center, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Qingyu Yuan
- Department of Medical Imaging Center, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Cheng Zhang
- Department of Gastrointestinal Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Kangneng Zhou
- University of Science and Technology, Beijing, China
| | - Zepang Sun
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Taojun Zhang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Zhen Han
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Hao Feng
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Xiaokun Liang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Shenzhen Colleges of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Yonghong Zhong
- Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haijun Deng
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Lequan Yu
- Department of Statistics and Actuarial Science, The University of Hong Kong, Hong Kong, China
| | - Yikai Xu
- Department of Medical Imaging Center, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Wei Wang
- Department of Gastrointestinal Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Guoxin Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Yuming Jiang
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
10
|
Ke L, Li S, Huang D. The predictive value of tumor mutation burden on survival of gastric cancer patients treated with immune checkpoint inhibitors: A systematic review and meta-analysis. Int Immunopharmacol 2023; 124:110986. [PMID: 37748223 DOI: 10.1016/j.intimp.2023.110986] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND Tumor mutation burden (TMB) is a complement to traditional biomarkers related to the efficacy of immune checkpoint inhibitors (ICIs). The relationship between TMB and the efficacy of ICIs in gastric cancer was controversial. The systematic review and meta-analysis were conducted to investigate the predictive value of TMB on survival of gastric cancer patients treated with ICIs. METHODS We searched the databases PubMed, Embase, and Web of Science for articles, then screened eligible articles according to inclusion criteria. The effective data were extracted to calculate the pooled effects of hazard ratio (HR) for overall survival (OS) and progression-free survival (PFS), then perform publication bias, sensitivity analysis, and subgroup analysis by STATA 16.0. RESULTS The high TMB patients showed significantly longer survival than the low TMB patients (OS: HR 0.65,95% CI 0.55, 0.77, p < 0.001; PFS: HR 0.51, 95% CI 0.33, 0.77, p = 0.001). In the Asian subgroup, patients with high TMB exhibited better prognosis compared to low TMB (OS: HR 0.56, 95% CI 0.43, 0.72, p < 0.001; PFS: HR 0.45, 95% CI 0.28, 0.72, p = 0.001). In the non-Asian subgroup, the survival benefit was observed to be skewed toward patients with high TMB, but it was not statistically significant (OS:HR 0.61, 95% CI 0.32, 1.16, p = 0.133; PFS:HR 0.68, 95% CI 0.31, 1.48, p = 0.322). CONCLUSIONS This meta-analysis demonstrated that gastric cancer patients with high TMB showed significant benefits from ICIs compared to those with low TMB patients, particularly in Asian populations.
Collapse
Affiliation(s)
- Liyuan Ke
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China.
| | - Su Li
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Danxue Huang
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
11
|
Takayama T, Tsuji Y. Updated Adjuvant Chemotherapy for Gastric Cancer. J Clin Med 2023; 12:6727. [PMID: 37959193 PMCID: PMC10648766 DOI: 10.3390/jcm12216727] [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: 08/25/2023] [Revised: 10/03/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023] Open
Abstract
Surgical resection is currently the best curative approach for gastric cancer (GC); however, the prognosis of patients with advanced GC remains poor even with curative resection. For this reason, perioperative chemotherapy has been combined with surgery to reduce the risk of postoperative recurrence. Standard perioperative chemotherapy for resectable advanced GC varies from region to region. Postoperative S-1 therapy was standardized via the ACTS-GC study in East Asia, perioperative ECF (Epirubicin + Cisplatin + Fluorouracil) was standardized via the MAGIC study in Europe, and postoperative chemoradiotherapy was standardized via the US intergroup study in North America. Since then, more intensive regimens have been developed. In recent years, perioperative therapy using novel agents, such as molecular-targeted drugs and immune checkpoint inhibitors (ICIs), has also been tested and evaluated in the three major regions (East Asia, Europe, and North America) with promising results. Perioperative chemotherapy has become an integral part of many treatment strategies and requires continued research and evaluation.
Collapse
Affiliation(s)
- Toshizo Takayama
- Department of Medical Oncology, Tonan Hospital, Sapporo 060-0004, Japan
- Department of Medical Oncology, Daido Hospital, Nagoya 457-8511, Japan
| | - Yasushi Tsuji
- Department of Medical Oncology, Tonan Hospital, Sapporo 060-0004, Japan
| |
Collapse
|
12
|
Lu M, Wu Y, Zhang Y, Yu Y, Wang S, Su X. Immunotherapeutic strategy in the management of gastric cancer: molecular profiles, current practice, and ongoing trials. J Egypt Natl Canc Inst 2023; 35:32. [PMID: 37779128 DOI: 10.1186/s43046-023-00192-1] [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: 08/25/2023] [Accepted: 09/15/2023] [Indexed: 10/03/2023] Open
Abstract
Gastric cancer (GC) is the one of the most commonly solid cancer worldwide. Although under the aggressive treatment, the poor clinical outcomes of patients with GCs have not been improved. Current studies emphasized that targeting therapies or immune response-based therapeutic strategy may be a potential approach to improve the clinical outcomes. Moreover, accumulative evidence has reported the increasing expression of PD-L1 expression in GC cells and highlighted its role in the tumor progression. Currently, great development has been established in the immune checkpoint inhibitors (ICIs) and further changed the clinical practice of GC treatment and prognosis. In addition, the combination therapies with targeting therapy or traditional therapies are expected to push the development of immunotherapies. In our present review, we predominantly focus on the biomarkers and molecular profiles for immunotherapies in GCs and highlight the role and administration of ICIs-based immunotherapeutic strategies against the GCs.
Collapse
Affiliation(s)
- Mengxiao Lu
- Department of Gastrointestinal Minimally Invasive Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China.
| | - Yingjie Wu
- Department of Gastrointestinal Minimally Invasive Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Yixin Zhang
- Department of Gastrointestinal Minimally Invasive Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Yu Yu
- Department of Gastrointestinal Minimally Invasive Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | | | - Xiaobao Su
- Department of Gastrointestinal Minimally Invasive Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
13
|
Dai X, Ding W, He Y, Huang S, Liu Y, Wu T. Refractory microsatellite stable metastatic colorectal cancer with ERBB2/ERBB3 mutation may be preferred population for regorafenib plus PD-1 inhibitor therapy: a real-world study. Front Oncol 2023; 13:1227644. [PMID: 37681031 PMCID: PMC10482431 DOI: 10.3389/fonc.2023.1227644] [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: 05/23/2023] [Accepted: 08/08/2023] [Indexed: 09/09/2023] Open
Abstract
Background Microsatellite stable (MSS) colorectal cancer (CRC) has been referred to as the "cold tumor" because of almost no response to anti-programmed death-1 (PD-1) antibody. A recent REGONIVO trial showed that regorafenib plus nivolumab had an encouraging efficacy in MSS metastatic CRC (mCRC). However, only a small subset of patients may benefit from the combination therapy. We aim to evaluate the efficacy and safety data of immune checkpoint inhibitors combined with regorafenib in refractory MSS mCRC and to discover biomarkers that can effectively stratify the beneficial patient population. Methods We retrospectively analyzed patients with MSS mCRC who received regorafenib combined with anti-PD-1 antibody therapy. The objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and status of gene mutation were reviewed and evaluated. Results Twenty-one patients received combination treatment. At a median treatment duration of 4 months, one patient achieved complete response, three patients achieved partial response, and two patients achieved stable disease as the best response. The ORR and DCR were 19% and 28.5% in the overall population, respectively. The median PFS was 4 months, and the median OS was 25 months. Only erbb2 receptor tyrosine kinase 2/erbb3 receptor tyrosine kinase 3 (ERBB2/ERBB3) mutation status was confirmed to be a potential predictive factor for effective treatment. In patients with ERBB2/ERBB3 mutation, ORR, DCR, and PFS exhibited significant improvements in comparison with that in wild-type patients. Grade 3 or higher treatment-related adverse events occurred in three patients (14.3%). Conclusions Regorafenib in combination with PD-1 inhibitor provides a feasible treatment regimen for refractory MSS mCRC with tolerated toxicity. Patients with ERBB2/ERBB3 mutation may be the preferred population for this combination regimen.
Collapse
Affiliation(s)
| | | | | | | | - Yun Liu
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingyu Wu
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Zhou ML, Xu RN, Tan C, Zhang Z, Wan JF. Advanced gastric cancer achieving major pathologic regression after chemoimmunotherapy combined with hypofractionated radiotherapy: A case report. World J Gastrointest Oncol 2023; 15:1096-1104. [PMID: 37389115 PMCID: PMC10302995 DOI: 10.4251/wjgo.v15.i6.1096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/09/2023] [Accepted: 04/23/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Currently, chemotherapy combined with immunotherapy is the established first-line standard treatment for advanced gastric cancer (GC). In addition, the combination of radiotherapy and immunotherapy is considered a promising treatment strategy.
CASE SUMMARY In this report, we present a case of achieving nearly complete remission of highly advanced GC with comprehensive therapies. A 67-year-old male patient was referred to the hospital because he presented with dyspepsia and melena for several days. Based on fluorodeoxyglucose positron emission tomography/computed tomography (FDG PET/CT), endoscopic examination and abdominal CT, he was diagnosed with GC with a massive lesion and two distant metastatic lesions. The patient received mFOLFOX6 regimen chemotherapy, nivolumab and a short course of hypofractionated radiotherapy (4 Gy × 6 fractions) targeting the primary lesion. After the completion of these therapies, the tumor and the metastatic lesions showed a partial response. After having this case discussed by a multidisciplinary team, the patient underwent surgery, including total gastrectomy and D2 lymph node dissection. Postoperative pathology showed that major pathological regression of the primary lesion was achieved. Chemoimmunotherapy started four weeks after surgery, and examination was performed every three months. Since surgery, the patient has been stable and healthy with no evidence of recurrence.
CONCLUSION The combination of radiotherapy and immunotherapy for GC is worthy of further exploration.
Collapse
Affiliation(s)
- Meng-Long Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University, Shanghai 200032, China
| | - Ruo-Ne Xu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University, Shanghai 200032, China
| | - Cong Tan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University, Shanghai 200032, China
| | - Jue-Feng Wan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University, Shanghai 200032, China
| |
Collapse
|
15
|
Jiang Q, Liu W, Zeng X, Zhang C, Du Y, Zeng L, Yin Y, Fan J, Yang M, Tao K, Zhang P. Safety and efficacy of tislelizumab plus chemotherapy versus chemotherapy alone as neoadjuvant treatment for patients with locally advanced gastric cancer: real-world experience with a consecutive patient cohort. Front Immunol 2023; 14:1122121. [PMID: 37215127 PMCID: PMC10195027 DOI: 10.3389/fimmu.2023.1122121] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Objectives Immunotherapy plus chemotherapy has recently been applied in the neoadjuvant treatment for locally advanced gastric cancer (LAGC), while its superiority over neoadjuvant chemotherapy (NACT) alone remains to be explored. This study explored the safety and efficacy of NACT plus tislelizumab in patients with LAGC. Methods The data on patients with LAGC who received NACT combined with radical gastrectomy and NACT plus tislelizumab followed by radical gastrectomy was retrospectively collected. Clinicopathological characteristics of the two groups were compared. Results A total of 119 and 50 patients with gastric cancer treated with NACT and NACT plus tislelizumab, respectively, were enrolled. No significant difference was found between the baseline data of the two groups. The operative time (210.5 ± 70.4 min vs. 237.6 ± 68.4 min, P=0.732), intraoperative blood loss (157.8 ± 75.9 ml vs. 149.1 ± 92.5 ml, P=0.609), and number of dissected lymph nodes (24.7 ± 9.3 vs. 28.1 ± 10.3, P=0.195) was not statistically different between the two groups. In comparison to the NACT plus tislelizumab group, the R0 resection rate (100% vs. 89.9%, P=0.019) and pathologic complete response rate (26.0% vs. 3.4%, P<0.001) were significantly lower in the NACT group. The postoperative complication rates were 24.4% and 26.0% in the NACT and NACT plus tislelizumab groups with no significant difference (P=0.823). In subgroup analysis, tumor regression grade (TRG) (TRG 3: 72.3% vs. 23.5%, P<0.001) and ypN stage (stages 2-3: 46.8% vs. 5.9%, P=0.003) in the NACT group were significantly higher compared with the NACT plus tislelizumab group in esophagogastric junction carcinoma. Conclusion Compared with the S-1 and oxaliplatin (SOX) or 5-fluorouracil, folinic acid, and oxaliplatin (FOLFOX) NACT regimen, NACT plus tislelizumab significantly improved the efficacy and R0 resection rate of LAGC without increasing the incidence of perioperative complications, particularly in esophagogastric junction carcinoma.
Collapse
Affiliation(s)
- Qi Jiang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Weizhen Liu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiangyu Zeng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chenggang Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuqiang Du
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Liwu Zeng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuping Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Fan
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ming Yang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Peng Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
16
|
Racz K, Legner A, Böhme F, Sebesta C. [Gastric cancer]. Wien Med Wochenschr 2023; 173:227-231. [PMID: 37071301 DOI: 10.1007/s10354-023-01011-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/06/2023] [Indexed: 04/19/2023]
Abstract
Gastric cancers are relativ commonly cancer types. The therapy options have changed in the last years as well in the surgery as in the oncology, it is worth to look at the etiology, diagnosis and therapy.
Collapse
Affiliation(s)
- Krisztina Racz
- 2. Medizinische Abteilung, Klinik Donaustadt, Donaustadt, Österreich.
| | - Andras Legner
- Chirurgische Abteilung, Klinik Donaustadt, Donaustadt, Österreich
| | - Felix Böhme
- 2. Medizinische Abteilung, Klinik Donaustadt, Donaustadt, Österreich
| | - Christian Sebesta
- 2. Medizinische Abteilung, Klinik Donaustadt, Donaustadt, Österreich
| |
Collapse
|
17
|
Liu Y, Hu P, Xu L, Zhang X, Li Z, Li Y, Qiu H. Current Progress on Predictive Biomarkers for Response to Immune Checkpoint Inhibitors in Gastric Cancer: How to Maximize the Immunotherapeutic Benefit? Cancers (Basel) 2023; 15:2273. [PMID: 37190201 PMCID: PMC10137150 DOI: 10.3390/cancers15082273] [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: 03/20/2023] [Revised: 04/09/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Gastric cancer is the fifth most prevalent cancer and the fourth leading cause of cancer death globally. Delayed diagnosis and pronounced histological and molecular variations increase the complexity and challenge of treatment. Pharmacotherapy, which for a long time was systemic chemotherapy based on 5-fluorouracil, is the mainstay of management for advanced gastric cancer. Trastuzumab and programmed cell death 1 (PD-1) inhibitors have altered the therapeutic landscape, contributing to noticeably prolonged survivorship in patients with metastatic gastric cancer. However, research has revealed that immunotherapy is only beneficial to some individuals. Biomarkers, such as programmed cell death ligand 1 (PD-L1), microsatellite instability (MSI), and tumor mutational load (TMB), have been shown to correlate with immune efficacy in numerous studies and are increasingly employed for the selection of patients most likely to respond to immunotherapy. Gut microorganisms, genetic mutations like POLE/POLD1 and NOTCH4, tumor lymphoid infiltrating cells (TILs), and other novel biomarkers have the potential to develop into new predictors. Prospective immunotherapy for gastric cancer should be guided by a biomarker-driven precision management paradigm, and multidimensional or dynamic marker testing could be the way to go.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hong Qiu
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.L.)
| |
Collapse
|
18
|
Jiang H, Wang J, Deng W. Pathologic complete response to chemoimmunotherapy of an advanced gastric cancer patient with high PD-L1 expression, dMMR, and unique gut microbiota composition: A case report. Front Oncol 2023; 13:1150931. [PMID: 37007083 PMCID: PMC10061119 DOI: 10.3389/fonc.2023.1150931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
BackgroundAdvanced gastric cancer (AGC) is a malignant disease with limited therapeutic options and a poor prognosis. Recently, immune checkpoint inhibitors (ICIs), represented by inhibitors of programmed cell death 1 (PD-1)/programmed death-ligand 1 (PD-L1), have emerged as a potential gastric cancer (GC) therapy.Case presentationThis case study aimed to reveal the tumor response to neoadjuvant chemotherapy combined with camrelizumab in a patient with AGC based on the characteristics of the clinical pathology, genomics variation, and gut microbiome. Samples from a 59-year-old male patient diagnosed with locally advanced unresectable GC (cT4bN2M0, high grade) presenting PD-L1-positive, deficient mismatch repair (dMMR), and highly specific gut microbiota enrichment were subjected to target region sequencing, metagenomic sequencing, and immunohistochemistry staining. The patient received neoadjuvant therapy, including camrelizumab, apatinib, S-1, and abraxane, which eventually promoted dramatic tumor shrinkage without serious adverse effects and allowed subsequent radical gastrectomy and lymphadenectomy. Finally, the patient achieved pathologic complete response (pCR), and the recurrence-free survival time was 19 months at the last follow-up in April 2021.ConclusionsThe patient with PD-L1-positive, dMMR, and a highly specific gut microbiota enrichment exhibited a pCR to neoadjuvant chemoimmunotherapy.
Collapse
Affiliation(s)
- Hongpeng Jiang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Junyun Wang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
| | - Wei Deng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & National Clinical Research Center for Digestive Diseases, Beijing, China
- *Correspondence: Wei Deng,
| |
Collapse
|
19
|
Identification of Immunogenic Cell-Death-Related Subtypes and Development of a Prognostic Signature in Gastric Cancer. Biomolecules 2023; 13:biom13030528. [PMID: 36979463 PMCID: PMC10046021 DOI: 10.3390/biom13030528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023] Open
Abstract
Background: Immunogenic cell death (ICD) is considered a promising type of regulated cell death and exerts effects by activating the adaptive immune response, reshaping the tumor environment (TME) and improving therapeutic efficacy. However, the potential roles and prognostic value of ICD-associated genes in gastric cancer (GC) remain unclear. Methods: The RNA expression data and clinical information of 1090 GC patients from six cohorts were collected. Consensus clustering was used to identify three distinct molecular subtypes. Then, a robust prognostic ICD_score for predicting prognosis was built via WGCNA and LASSO Cox regression according to the TCGA cohort, and the predictive capability of the ICD_score in GC patients was validated in the other cohorts. ICD-related immune features were analyzed using a CIBERSORT method and verified by immunofluorescence. Results: We found that ICD-related gene variations were correlated with clinical outcomes, tumor immune microenvironment (TIME) characteristics and treatment response. We then constructed an ICD signature that classifies cases as low- and high-ICD_score groups. The high-ICD_score group indicates unfavorable OS, a more advanced TNM stage, and presents an immune-suppressed phenotype, which has more infiltrations of pro-tumor immune cells, such as macrophages, which was verified by immunofluorescence. In addition, a nomogram containing the ICD_score showed a high predictive accuracy with AUCs of 0.715, 0.731 and 0.8 on Years 1, 3, and 5. Conclusion: We performed the first and synthesis ICD analysis in GC and built a clinical application tool based on the ICD signature, which paved a new path for assessing prognosis and guiding individual treatment.
Collapse
|
20
|
Clinical relevance of PD-1 positive CD8 T-cells in gastric cancer. Gastric Cancer 2023; 26:393-404. [PMID: 36781556 PMCID: PMC10115710 DOI: 10.1007/s10120-023-01364-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 01/11/2023] [Indexed: 02/15/2023]
Abstract
BACKGROUND We evaluated the relevance of PD-1+CD8+ T-cells in gastric cancer (GC) including prognostic significance, association with chemotherapy and immunotherapy sensitivity and correlations with the tumor microenvironment (TME). METHODS Discovery cohort: GC samples were evaluated for AE1/3, CD8, PD-1, Ki-67 and Granzyme-B expression with fluorescence-based multiplex immunohistochemistry (mIHC). Validation cohorts: we analyzed bulk RNAseq GC datasets from TCGA, the "3G" chemotherapy trial and an immunotherapy phase 2 trial. The cox proportional hazards model was used to identify factors that influenced overall survival (OS). To study the TME, we analyzed single-cell RNAseq performed on GCs. RESULTS In the discovery cohort of 350 GCs, increased PD-1 expression of CD8 T-cells was prognostic for OS (HR 0.822, p = 0.042). PD-1 expression in CD8 T-cells highly correlated with cytolytic [Granzyme-B+] (r = 0.714, p < 0.001) and proliferative [Ki-67+] (r = 0.798, p < 0.001) activity. Analysis of bulk RNAseq datasets showed tumors with high PD-1 and CD8A expression levels had improved OS when treated with immunotherapy (HR 0.117, p = 0.036) and chemotherapy (HR 0.475, p = 0.017). Analysis of an scRNAseq dataset of 152,423 cells from 40 GCs revealed that T-cell and NK-cell proportions were higher (24% vs 18% and 19% vs 15%, p < 0.0001), while macrophage proportions were lower (7% vs 11%, p < 0.0001) in CD8PD-1high compared to CD8PD-1low tumors. CONCLUSION This is one of the largest GC cohorts of mIHC combined with analysis of multiple datasets providing orthogonal validation of the clinical relevance of PD-1+CD8+ T-cells being associated with improved OS. CD8PD-1high tumors have distinct features of an immunologically active, T-cell inflamed TME.
Collapse
|
21
|
Tang X, Li M, Wu X, Guo T, Zhang L, Tang L, Jia F, Hu Y, Zhang Y, Xing X, Shan F, Gao X, Li Z. Neoadjuvant PD-1 blockade plus chemotherapy induces a high pathological complete response rate and anti-tumor immune subsets in clinical stage III gastric cancer. Oncoimmunology 2022; 11:2135819. [PMID: 36268179 PMCID: PMC9578498 DOI: 10.1080/2162402x.2022.2135819] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
First-line PD-1 blockade plus chemotherapy significantly improves the survival benefits in late-stage gastric cancer (GC) patients. However, the pathological response rate and effects on the immune microenvironment of neoadjuvant PD-1 blockade plus chemotherapy in patients with cTNM-stage III GC remain to be elucidated. Patients with cTNM-stage III GC who underwent neoadjuvant PD-1 blockade plus chemotherapy and surgery were enrolled. Four in vivo models bearing GC were jointly established to investigate the specific roles of chemotherapy and PD-1 blockade for GC treatment. The tumor immune microenvironment was analyzed by hematoxylin and eosin (H&E) and IHC staining, multicolor flow cytometry and immunofluorescence. A total of 75 patients with cTNM-stage III (cT2-4N1-3M0) gastric cancer who received neoadjuvant PD-1 blockade plus chemotherapy (SOX/XELOX) were included in this study. After treatment, 21 (28.0%) and 57 (76.0%) patients achieved pathological complete response (pCR) and post-therapy pathological downstaging. Subgroup analyses revealed that patients with CPS >1 (32.6% vs 8.3%) and dMMR (35.7% vs 25.4%) subtype had better efficacy. Additionally, the resected specimens showed more anti-tumor immune infiltration indicating a response to neoadjuvant PD-1 blockade plus chemotherapy. Multicolor immunofluorescence and in vivo experiments on mouse models revealed that elevated M1/M2 ratio of macrophages, CD8 + T cells and plasma cells indicated effective response to treatment. Furthermore, neoadjuvant PD-1 blockade plus chemotherapy neither delayed surgery nor increased postoperative complication rate. The analyses indicate neoadjuvant PD-1 blockade plus chemotherapy is a promising therapeutic strategy in patients with cTNM-stage III GC with an encouraging pCR rate.
Collapse
Affiliation(s)
- Xiaohuan Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Mengyuan Li
- Department of Radiation Oncology, Peking University 3rd Hospital, Beijing, P.R. China
| | - Xiaolong Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Ting Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Li Zhang
- Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Lei Tang
- Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Fangzhou Jia
- Biological Sample Bank, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Ying Hu
- Biological Sample Bank, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Yan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Xiaofang Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Fei Shan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Xiangyu Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Ziyu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| |
Collapse
|
22
|
Zheng J, Huang B, Xiao L, Wu M, Li J. Treatment- and immune-related adverse events of immune checkpoint inhibitors in esophageal or gastroesophageal junction cancer: A network meta-analysis of randomized controlled trials. Front Oncol 2022; 12:821626. [PMID: 36568203 PMCID: PMC9780048 DOI: 10.3389/fonc.2022.821626] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 11/14/2022] [Indexed: 12/13/2022] Open
Abstract
Objective To systematically evaluate the safety and adverse event profiles of immune checkpoint inhibitors (ICIs) in patients with esophageal cancer (EPC) or gastroesophageal junction cancer (GEJC). Methods PubMed, Web of Science, Cochrane Library, and major conference proceedings were systematically searched for all phase II or phase III randomized controlled trials (RCTs) in EPC or GEJC using ICIs. Safety outcomes including treatment-related adverse events (trAEs), immune-related adverse events (irAEs), and serious trAEs were evaluated by network meta-analysis or dichotomous meta-analysis based on the random-effects model. Results Eleven RCTs involving EPC (five RCTs) and GEJC (six RCTs) were included in the final meta-analysis. NMA showed that placebo was associated with the best safety ranking for grade 3-5 trAEs (SUCRA = 96.0%), followed by avelumab (78.6%), nivolumab (73.9%), ipilimumab (57.0%), and pembrolizumab (56.6%). Conventional pairwise meta-analysis (CPM) showed that ICIs have similar grade 3-5 trAE risk compared with chemotherapy (RR = 0.764, 95% CI: 0.574 to 1.016, I 2 = 95.7%, Z = 1.85, P = 0.065). NMA showed that the general safety of grade 3-5 irAEs ranked from high to low is as follows: ChT (85.1%), placebo (76.5%), ipilimumab (56.0%), nivolumab (48.5%), avelumab (48.4%), camrelizumab (41.8%), pembrolizumab (36.4%), and nivolumab + ipilimumab (21.6%). CPM showed that the rates of grade 3-5 irAEs in the ICI group and the chemotherapy group were 7.35% (154/2,095, 95% CI: [6.23%, 8.47%]) versus 2.25% (42/1,869, 95% CI: [1.58%, 2.92%]), with statistical significance (RR = 3.151, 95% CI = 2.175 to 4.563, Z = 6.07, P = 0.000). The most common irAEs in the ICI group were skin reaction (15.76%, 95% CI: [13.67%, 17.84%]), followed by hypothyroidism (9.73%, 95% CI: [8.07%, 11.39%]), infusion-related reactions (5.93%, 95% CI: [4.29%, 7.58%]), hepatitis (5.25%, 95% CI: [4.28%, 6.22%]), and pneumonitis (4.45%, 95% CI: [3.5%, 5.4%]). Conclusion Different ICIs had different toxicity manifestations and should not be considered as an entity. Compared with chemotherapy, ICIs were more prone to irAEs, but the overall rates remained low and acceptable. For clinicians, it is important to recognize and monitor the adverse events caused by ICIs for patients with EPC or GEJC.
Collapse
Affiliation(s)
- Jianqing Zheng
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China,*Correspondence: Jiancheng Li, ; Jianqing Zheng,
| | - Bifen Huang
- Department of Obstetrics and Gynecology, Quanzhou Medical College People’s Hospital Affiliated, Quanzhou, China
| | - Lihua Xiao
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Min Wu
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Jiancheng Li
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China,*Correspondence: Jiancheng Li, ; Jianqing Zheng,
| |
Collapse
|
23
|
Wang J, Tu S, Chavda VP, Chen ZS, Chen X. Successes and failures of immunotherapy for gastric cancer. Drug Discov Today 2022; 27:103343. [PMID: 36075377 DOI: 10.1016/j.drudis.2022.103343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 07/13/2022] [Accepted: 08/31/2022] [Indexed: 11/03/2022]
Abstract
Many exploratory clinical studies have been conducted on immune checkpoint inhibitors (ICIs) as new therapeutic approaches for the first-line treatment of patients with advanced gastric cancer. Despite varying interpretations of the successes and failures of this clinical research, most analyses have focused on the results from the perspective of exploring the superiority of immunotherapy. Consequently, the role of chemotherapy as an important partner of immunotherapy in first-line combination therapy regimens for gastric cancer has attracted less attention. Here, we explore and analyze first-line immunotherapies for gastric cancer from the perspective of chemotherapy, to understand reasons for the failure of studies and to indicate directions for future clinical research.
Collapse
Affiliation(s)
- Jianzheng Wang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Shuiping Tu
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St John's University, Queens, NY 11439, USA.
| | - Xiaobing Chen
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China.
| |
Collapse
|
24
|
Wan M, Ding Y, Mao C, Ma X, Li N, Xiao C, Qian J, Jiang H, Zheng Y, Wu L, Teng L, Xu N. Association of inflammatory markers with survival in patients with advanced gastric cancer treated with immune checkpoint inhibitors combined with chemotherapy as first line treatment. Front Oncol 2022; 12:1029960. [PMID: 36387183 PMCID: PMC9650180 DOI: 10.3389/fonc.2022.1029960] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/17/2022] [Indexed: 12/27/2023] Open
Abstract
BACKGROUND The emergence of immune checkpoint inhibitors has changed the landscape of first-line treatment of patients with advanced gastric cancer. Currently, the prognostic significance of inflammatory markers in first-line immunotherapy combined with chemotherapy for gastric cancer is currently unclear. This study aimed to identify inflammatory markers with potential to predict treatment outcome in advanced gastric cancer patients receiving immunotherapy combined with chemotherapy. METHODS This retrospective study enrolled untreated advanced or metastatic gastric or gastro-esophageal junction cancer patients from 5 clinical trials (the clinical trial cohort) and the real world (the real-word cohort). Inflammatory markers included in the analysis included neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR), platelet-to-lymphocyte ratio (PLR), systemic inflammation index (SII), and derived neutrophil-to-lymphocyte ratio (dNLR). Receiver operating characteristic (ROC) curves were constructed to identify optimal cut-off values. The prognostic potential of the markers was determined using Kaplan-Meier analysis, univariate and multivariate Cox-regression analyses in the clinical trial cohort and the findings were validated in the real-world cohort. RESULTS In the clinical trial cohort (n=45), MLR, PLR and SII were associated with PFS but not OS (All P<0.05), while dNLR was not correlated with PFS or OS. Only NLR was associated with PFS and OS and identified as an independent prognostic predictor in the univariate and multivariate analyses. The prognostic value of NLR was validated in the real-world cohort (n=55). CONCLUSIONS NLR was a strong predictor of PFS and OS in patients with advanced gastric cancer receiving immune checkpoint inhibitors combined with chemotherapy. Further prospective studies are required to validate our results.
Collapse
Affiliation(s)
- Mingyu Wan
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Yongfeng Ding
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Chenyu Mao
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Xiaolu Ma
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Ning Li
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Cheng Xiao
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Jiong Qian
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Haiping Jiang
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Yulong Zheng
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Luntao Wu
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Lisong Teng
- Department of Surgical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| |
Collapse
|
25
|
Ma S, Chen F. Common strategies for effective immunotherapy of gastroesophageal cancers using immune checkpoint inhibitors. Pathol Res Pract 2022; 238:154110. [PMID: 36155325 DOI: 10.1016/j.prp.2022.154110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 11/21/2022]
Abstract
Gastroesophageal cancers (GECs) are very prevalent around the world and rank as the second cause of all cancer-related deaths in men and women and demonstrate a very poor prognosis. Currently, the treatment options for these malignancies are very limited and the response rates are also very low. Recently, immune checkpoint inhibitors (ICIs) have been proposed for immunotherapy of GECs; although preliminary results obtained from the clinical trials of ICIs in GECs were promising, they have shown to be effective only in a few subsets of patients who had a previous immune response to the tumor. In order to maximize the efficacy of ICIs in GECs, as well as identify the patients who will likely benefit from ICIs, several predictive biomarkers, such as Programmed death-ligand 1 (PD-L1) have been developed and evaluated. Since the single ICI therapies resulted in poor treatment response, several clinical studies began to explore various combinations of one or two ICIs with other anti-cancer treatment approaches, including chemotherapy, radiotherapy, and anti-angiogenesis therapy. These combinations demonstrated a more effective response among the ICIs-responsive patients and even in some instances sensitized the non-responsive individuals. This review is aimed to summarize the efforts made so far for improving the effectiveness of ICIs in the treatment of patients with GECs. Furthermore, multiple aspects of translational medicine such as available biomarkers and interactions between tumor and the immune system, as well as clinical aspects regarding the combination therapies and results of clinical trials will be discussed.
Collapse
Affiliation(s)
- Shuang Ma
- Cancer Center, Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, China.
| | - Fei Chen
- Department of Gastroenterology, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang People's Hospital), Taizhou 317200, China.
| |
Collapse
|
26
|
Camera S, Liscia N, Foti S, Barbieri L, Cossu A, Puccetti F, Elmore U, Rosati R, Scartozzi M, Mazza E, Cascinu S. Does immunotherapy change the treatment paradigm in metastatic gastric cancer? Med Oncol 2022; 39:224. [PMID: 36175723 DOI: 10.1007/s12032-022-01819-4] [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: 06/12/2022] [Accepted: 08/07/2022] [Indexed: 06/16/2023]
Abstract
Gastric cancer represents one of the leading causes of cancer-related death worldwide. Even if the last decade has witnessed an improvement in surgical and systemic treatments, with an increase of overall life expectancy, survival rates still remain unsatisfactory, especially for patients with metastatic disease. Systemic therapies represent the gold standard in the management of stage IV gastric cancer. In this scenario, the availability of effective second and third lines has represented for a long time the only hope to offer an overall survival improvement to these patients. Recently, the advent of immune checkpoint inhibitors has involved also gastric cancer with encouraging efficacy data in the metastatic setting, becoming integral part of the management of selected patients.
Collapse
Affiliation(s)
- Silvia Camera
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132, Milan, Italy.
| | - Nicole Liscia
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - Silvia Foti
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - Lavinia Barbieri
- Gastrointestinal Surgery Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - Andrea Cossu
- Gastrointestinal Surgery Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - Francesco Puccetti
- Gastrointestinal Surgery Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - Ugo Elmore
- Gastrointestinal Surgery Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - Riccardo Rosati
- Gastrointestinal Surgery Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042, Cagliari, Italy
| | - Elena Mazza
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - Stefano Cascinu
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132, Milan, Italy
| |
Collapse
|
27
|
Gou M, Qian N, Zhang Y, Wei L, Fan Q, Wang Z, Dai G. Construction of a nomogram to predict the survival of metastatic gastric cancer patients that received immunotherapy. Front Immunol 2022; 13:950868. [PMID: 36225924 PMCID: PMC9549034 DOI: 10.3389/fimmu.2022.950868] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Background Immunotherapy has shown promising results for metastatic gastric cancer (MGC) patients. Nevertheless, not all patients can benefit from anti-PD-1 treatment. Thus, this study aimed to develop and validate a prognostic nomogram for MGC patients that received immunotherapy. Methods Herein, MGC patients treated with anti-PD-1 between 1 October 2016 and 1 June 2022 at two separate Chinese PLA General Hospital centers were enrolled and randomly divided into training and validation sets (186 and 80 patients, respectively). The nomogram was constructed based on a multivariable Cox model using baseline variables from the training cohort. Its predictive accuracy was validated by the validation set. The consistency index (C-index) and calibration plots were used to evaluate the discriminative ability and accuracy of the nomogram. The net benefit of the nomogram was evaluated using decision curve analysis (DCA). Finally, we stratified patients by median total nomogram scores and performed Kaplan–Meier survival analyses. Results We developed the nomogram based on the multivariate analysis of the training cohort, including four parameters: surgery history, treatment line, lung immune prognostic index (LIPI), and platelet-to-lymphocyte ratio (PLR). The C-index of the nomogram was 0.745 in the training set. The calibration curve for 1- and 2-year survival showed good agreement between nomogram predictions and actual observations. In the validation group, the calibration curves demonstrated good performance of the nomogram, with a C-index for overall survival (OS) prediction of 0.713. The OS of patients with a score greater than the median nomogram score was significantly longer than patients with a score lower or equal to the median (p < 0.001). Conclusion We constructed a nomogram to predict the outcomes of MGC patients that received immunotherapy. This nomogram might facilitate individualized survival predictions and be helpful during clinical decision-making for MGC patients under anti-PD-1 therapy.
Collapse
Affiliation(s)
- Miaomiao Gou
- Medical Oncology Department, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Niansong Qian
- Medical Oncology Department, Hainan Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Yong Zhang
- Medical Oncology Department, The Second Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Lihui Wei
- Department of Medicine, Genetron Health (Beijing) Co. Ltd., Beijing, China
| | - Qihuang Fan
- Department of Medicine, Genetron Health (Beijing) Co. Ltd., Beijing, China
| | - Zhikuan Wang
- Medical Oncology Department, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
- *Correspondence: Guanghai Dai, ; Zhikuan Wang,
| | - Guanghai Dai
- Medical Oncology Department, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
- *Correspondence: Guanghai Dai, ; Zhikuan Wang,
| |
Collapse
|
28
|
Morgan E, Soerjomataram I, Rumgay H, Coleman HG, Thrift AP, Vignat J, Laversanne M, Ferlay J, Arnold M. The Global Landscape of Esophageal Squamous Cell Carcinoma and Esophageal Adenocarcinoma Incidence and Mortality in 2020 and Projections to 2040: New Estimates From GLOBOCAN 2020. Gastroenterology 2022; 163:649-658.e2. [PMID: 35671803 DOI: 10.1053/j.gastro.2022.05.054] [Citation(s) in RCA: 466] [Impact Index Per Article: 155.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 05/18/2022] [Accepted: 05/30/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS The aim of this study was to provide an overview of the burden of esophageal cancer in 185 countries in 2020 and projections for the year 2040. METHODS Estimates of esophageal cancer cases and deaths were extracted from the GLOBOCAN database for 2020. Age-standardized incidence and mortality rates were calculated overall, by sex, histologic subtype (adenocarcinoma [AC] and squamous cell carcinoma [SCC]), country, and level of human development for 185 countries. The predicted burden of incidence and mortality in 2040 was calculated based on global demographic projections. RESULTS Globally, there were an estimated 604,100 new cases of, and 544,100 deaths from, esophageal cancer in 2020, corresponding to age-standardized incidence and mortality rates of 6.3 and 5.6 per 100,000, respectively. Most cases were SCCs (85% [512,500 cases]) and 14% (85,700 cases) were ACs. Incidence and mortality rates were 2- to 3-fold higher in male (9.3 and 8.2, respectively) compared with female (3.6 and 3.2, respectively) individuals. Global variations in incidence and mortality were observed across countries and world regions; the highest rates occurred in Eastern Asia and Southern and Eastern Africa and the lowest occurred in Western Africa and Central America regions. If rates remain stable, 957,000 new cases (141,300 AC cases and 806,000 SCC cases) and 880,000 deaths from esophageal cancer are expected in 2040. CONCLUSIONS These updated estimates of the global burden of esophageal cancer represent an important baseline for setting priorities in policy making and developing and accelerating cancer control initiatives to reduce the current and projected burden. Although primary prevention remains key, screening and early detection represent important components of esophageal cancer control in high-risk populations.
Collapse
Affiliation(s)
- Eileen Morgan
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France.
| | | | - Harriet Rumgay
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Helen G Coleman
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK; Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Aaron P Thrift
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, Texas; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Jérôme Vignat
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Mathieu Laversanne
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Jacques Ferlay
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Melina Arnold
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| |
Collapse
|
29
|
Smyth E, Cozens K, Griffiths D, Clark KL, Ewings S, Petty R, Underwood T, Fitzgerald RC, Tanner J, Giger O, Anand S, Griffiths G. ELEVATE - evaluating Temozolomide and Nivolumab in patients with advanced unresectable previously treated oesophagogastric adenocarcinoma with MGMT methylation: study protocol for a single arm phase II trial. BMC Cancer 2022; 22:946. [PMID: 36050653 PMCID: PMC9434527 DOI: 10.1186/s12885-022-09891-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/13/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND For patients with oesophagogastric adenocarcinoma, surgery is the only curative option and despite the use of multimodality therapy, which combines it with chemotherapy and/or radiotherapy, more than 50% of patients will relapse and die. Many UK patients present with advanced disease which is already inoperable or metastatic at diagnosis. For these patients, standard care chemotherapy only offers them survival of less than a year. Nivolumab, a checkpoint blockade inhibitor, has been found to work in some advanced cancers. It is proposed, for those where immunotherapy hasn't worked, that these immunologically evasive tumours need to be sensitized to immunotherapy drugs to allow them to act. METHODS ELEVATE is a single arm phase II trial testing the overall response to nivolumab following temozolomide treatment in patients with advanced unresectable previously treated adenocarcinoma which is O6-methylguanine-DNA-methyltransferase (MGMT) methylated. 18 patients are being recruited from UK secondary care sites. To be eligible, participants must have been treated with at least 3 months of platinum and fluoropyrimidine chemotherapy. Participants will receive 50 mg/m2 temozolomide continuously for 3 months. If their disease progresses during the 3 months, they will stop temozolomide and start nivolumab at a dose of 240mg every 2 weeks. If there is no progression after 3 months the participant will continue taking temozolomide in combination with nivolumab. All treatment will stop once the participant progresses on nivolumab. The primary endpoint is the best overall response to nivolumab, using both Response Evaluation Criteria in Solid Tumours version 1.1 and immunotherapy modified Response Evaluation Criteria in Solid Tumours. Secondary endpoints include progression-free survival, overall survival, and quality of life. DISCUSSION ELEVATE will provide evidence for whether giving nivolumab after temozolomide in patients with previously treated advanced oesophagogastric adenocarcinoma is safe and biologically effective prior to future randomised trials. TRIAL REGISTRATIONS EudraCT Number: 2020-004771-41 (issued 01 October 2020); ISCRTN11398887 (registered 14 July 2021).
Collapse
Affiliation(s)
- Elizabeth Smyth
- Cambridge University Hospitals National Health Service Foundation Trust, Hill's Road, Cambridge, UK
| | - Kelly Cozens
- Southampton Clinical Trials Unit, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Daniel Griffiths
- Southampton Clinical Trials Unit, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK.
| | - Kathryn L Clark
- Southampton Clinical Trials Unit, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Sean Ewings
- Southampton Clinical Trials Unit, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - Tim Underwood
- School of Cancer Sciences, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - James Tanner
- Cambridge University Hospitals National Health Service Foundation Trust, Hill's Road, Cambridge, UK
| | - Olivier Giger
- Cambridge University Hospitals National Health Service Foundation Trust, Hill's Road, Cambridge, UK
| | | | - Gareth Griffiths
- Southampton Clinical Trials Unit, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
30
|
Huang W, Jiang Y, Xiong W, Sun Z, Chen C, Yuan Q, Zhou K, Han Z, Feng H, Chen H, Liang X, Yu S, Hu Y, Yu J, Chen Y, Zhao L, Liu H, Zhou Z, Wang W, Wang W, Xu Y, Li G. Noninvasive imaging of the tumor immune microenvironment correlates with response to immunotherapy in gastric cancer. Nat Commun 2022; 13:5095. [PMID: 36042205 PMCID: PMC9427761 DOI: 10.1038/s41467-022-32816-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 08/17/2022] [Indexed: 12/24/2022] Open
Abstract
The tumor immune microenvironment (TIME) is associated with tumor prognosis and immunotherapy response. Here we develop and validate a CT-based radiomics score (RS) using 2272 gastric cancer (GC) patients to investigate the relationship between the radiomics imaging biomarker and the neutrophil-to-lymphocyte ratio (NLR) in the TIME, including its correlation with prognosis and immunotherapy response in advanced GC. The RS achieves an AUC of 0.795-0.861 in predicting the NLR in the TIME. Notably, the radiomics imaging biomarker is indistinguishable from the IHC-derived NLR status in predicting DFS and OS in each cohort (HR range: 1.694-3.394, P < 0.001). We find the objective responses of a cohort of anti-PD-1 immunotherapy patients is significantly higher in the low-RS group (60.9% and 42.9%) than in the high-RS group (8.1% and 14.3%). The radiomics imaging biomarker is a noninvasive method to evaluate TIME, and may correlate with prognosis and anti PD-1 immunotherapy response in GC patients.
Collapse
Affiliation(s)
- Weicai Huang
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yuming Jiang
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Wenjun Xiong
- Department of Gastrointestinal Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road No. 111, Guangzhou, 510120, China
| | - Zepang Sun
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Chuanli Chen
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, China
| | - Qingyu Yuan
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, China
| | - Kangneng Zhou
- School of Computer and Communication Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Zhen Han
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Hao Feng
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Hao Chen
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Xiaokun Liang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Shitong Yu
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yanfeng Hu
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yan Chen
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liying Zhao
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Hao Liu
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhiwei Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, P. R. China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, P. R. China
| | - Wei Wang
- Department of Gastrointestinal Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road No. 111, Guangzhou, 510120, China.
| | - Wei Wang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, P. R. China.
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, P. R. China.
| | - Yikai Xu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, China.
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
31
|
Patel MA, Kratz JD, Lubner SJ, Loconte NK, Uboha NV. Esophagogastric Cancers: Integrating Immunotherapy Therapy Into Current Practice. J Clin Oncol 2022; 40:2751-2762. [PMID: 35839430 PMCID: PMC10476757 DOI: 10.1200/jco.21.02500] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 04/15/2022] [Accepted: 05/18/2022] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy (IO) agents have led to significant improvements in patient outcomes across many tumor types. There have been great efforts to introduce immune checkpoint inhibitors into the treatment paradigm of esophagogastric cancers as well. A number of randomized phase III trials, which will be reviewed here, established the role of these agents in both early-stage and advanced-stage disease. Adjuvant nivolumab is US Food and Drug Administration-approved after neoadjuvant chemoradiation and resection of esophageal and gastroesophageal junction cancers on the basis of the phase III CheckMate 577 trial. In the advanced setting, patients with programmed death receptor ligand-1-positive tumors should be recommended IO in combination with chemotherapy in the first-line setting on the basis of the results from KEYNOTE 590, CheckMate 649, and CheckMate 648. Across trials, chemotherapy continues to play a critical role in the first-line setting and should be offered to all patients who are eligible for systemic therapy, including those with biomarker select tumors. In the later lines of treatment, IO has modest activity, and prior studies have grown largely irrelevant because of the enrollment of IO-naive patients. Similar to other disease types, patients with microsatellite unstable (microsatellite instability high) tumors represent a unique cohort that is more sensitive to IO. However, there are no randomized studies evaluating how best to apply IO in early or advanced stages specifically for the treatment of patients with microsatellite instability high upper GI tumors. Questions remain how to best select patients who benefit from IO treatments, how to augment IO activity in programmed death receptor ligand-1-negative tumors, and how to incorporate IO in late-line settings or for recurrent disease that has been treated with IO-containing regimens during early stages.
Collapse
Affiliation(s)
- Monica A. Patel
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin, Madison, WI
- Carbone Cancer Center, Madison, WI
| | - Jeremy D. Kratz
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin, Madison, WI
- Carbone Cancer Center, Madison, WI
| | - Sam J. Lubner
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin, Madison, WI
- Carbone Cancer Center, Madison, WI
| | - Noelle K. Loconte
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin, Madison, WI
- Carbone Cancer Center, Madison, WI
| | - Nataliya V. Uboha
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin, Madison, WI
- Carbone Cancer Center, Madison, WI
| |
Collapse
|
32
|
Myer NM, Shitara K, Chung HC, Lordick F, Kelly RJ, Szabo Z, Cao ZA, Leong S, Ilson DH, Weichert W. Evolution of predictive and prognostic biomarkers in the treatment of advanced gastric cancer. J Cancer Res Clin Oncol 2022; 148:2023-2043. [PMID: 35551464 PMCID: PMC11110882 DOI: 10.1007/s00432-021-03902-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/24/2021] [Indexed: 12/30/2022]
Abstract
Despite new therapeutic options, advanced gastric cancer remains associated with a poor prognosis compared with other cancers. Recent gains in the treatment of gastric cancer were accompanied by the identification of novel biomarkers associated with various cellular pathways and corresponding diagnostic technologies. It is expected that the standardization of clinical workflow and technological refinements in biomarker assessment will support greater personalization and further improve treatment outcomes. In this article, we review the current state of prognostic and predictive biomarkers in gastric cancer.
Collapse
Affiliation(s)
- Nicole M Myer
- Merck & Co., Inc., 90 E. Scott Avenue, Rahway, NJ, 07065, USA.
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Hyun C Chung
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Florian Lordick
- Medical Department (Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases), University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, Leipzig, Germany
| | - Ronan J Kelly
- Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
| | - Zsolt Szabo
- Merck & Co., Inc., Ringstrasse 27 Kriens, LUZERN, 6010, Switzerland
| | - Z Alexander Cao
- Merck & Co., Inc., 90 E. Scott Avenue, Rahway, NJ, 07065, USA
| | - Stephen Leong
- Merck & Co., Inc., 351 N Sumneytown Pike, North Wales, PA, 19454, USA
| | - David H Ilson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wilko Weichert
- Institute of Pathology, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
33
|
Kumar S, Chatterjee M, Ghosh P, Ganguly KK, Basu M, Ghosh MK. Targeting PD-1/PD-L1 in cancer immunotherapy: an effective strategy for treatment of triple-negative breast cancer (TNBC) patients. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022] Open
|
34
|
Wang TY, Chen CY, Huang TH, Yang YH, Chen KJ, Chou WC, Lu CH. Protein-bound polysaccharide K prolonged overall survival in gastric cancer patients from a non-Japanese Asian country who received gastrectomy and adjuvant chemotherapy. Medicine (Baltimore) 2022; 101:e29632. [PMID: 35866836 PMCID: PMC9302315 DOI: 10.1097/md.0000000000029632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 04/19/2022] [Accepted: 05/06/2022] [Indexed: 01/17/2023] Open
Abstract
Adding protein-bound polysaccharide K (PSK) to adjuvant chemotherapy with mitomycin and fluorouracil after gastrectomy for gastric cancer was demonstrated to improve survival in a previous study in Japan. However, the efficacy of PSK outside Japan and in combination with other adjuvant chemotherapeutic agents remains unclear. The aims of this study were to evaluate the efficacy of PSK. We conducted a population-based historical cohort study using the National Health Insurance Research Database of Taiwan. We performed sensitivity analysis with propensity score matching to control for possible confounders. Patients who used PSK (PSK group) were matched at a 1:4 ratio to those who had never used PSK (control group) after adjusting for covariates including sex, age, urbanization, income and comorbidities. The primary outcome was overall survival. Multivariate hazard ratios from competing risk analysis were calculated by adjusting for demographic data and all confounding factors. From 1999 to 2008, we identified 10,617 patients with gastric cancer received gastrectomy and adjuvant chemotherapy. 1295 patients used PSK (PSK group) and 5180 patients never used PSK (control group) were analyzed after propensity score matching. The median overall survival was 6.49 years (95% confidence interval [CI] 5.22-7.63) in the PSK group and 3.59 years (95% CI 3.38-3.80) in the control group. After adjusting for age, sex, urbanization, income, and comorbidities, adding PSK to adjuvant chemotherapy was the most significant prognostic factor for improved survival (hazard ratio 0.76, P < .0001). Adjuvant chemotherapy combined with PSK significantly prolonged overall survival in gastric cancer patients after gastrectomy.
Collapse
Affiliation(s)
- Ting-Yao Wang
- Division of Hematology and Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Chao-Yu Chen
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Chiayi, Taiwan; Department of Early Childhood Care and Education, Shu-Zen Junior College of Medicine and Management, Kaohsiung, Taiwan
| | - Tzu-Hao Huang
- Division of General Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yao-Hsu Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan; School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Health Information and Epidemiology Laboratory, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Ko-Jung Chen
- Health Information and Epidemiology Laboratory, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Wen-Chi Chou
- Department of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou and Chang Gung University, College of Medicine, Taoyuan, Taiwan
| | - Chang-Hsien Lu
- Division of Hematology and Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| |
Collapse
|
35
|
Mukhopadhyay P, Ye J, Anderson KM, Roychoudhury S, Rubin EH, Halabi S, Chappell RJ. Log-Rank Test vs MaxCombo and Difference in Restricted Mean Survival Time Tests for Comparing Survival Under Nonproportional Hazards in Immuno-oncology Trials: A Systematic Review and Meta-analysis. JAMA Oncol 2022; 8:1294-1300. [PMID: 35862037 PMCID: PMC9305601 DOI: 10.1001/jamaoncol.2022.2666] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance The log-rank test is considered the criterion standard for comparing 2 survival curves in pivotal registrational trials. However, with novel immunotherapies that often violate the proportional hazards assumptions over time, log-rank can lose power and may fail to detect treatment benefit. The MaxCombo test, a combination of weighted log-rank tests, retains power under different types of nonproportional hazards. The difference in restricted mean survival time (dRMST) test is frequently proposed as an alternative to the log-rank under nonproportional hazard scenarios. Objective To compare the log-rank with the MaxCombo and dRMST in immuno-oncology trials to evaluate their performance in practice. Data Sources Comprehensive literature review using Google Scholar, PubMed, and other sources for randomized clinical trials published in peer-reviewed journals or presented at major clinical conferences before December 2019 assessing efficacy of anti-programmed cell death protein-1 or anti-programmed death/ligand 1 monoclonal antibodies. Study Selection Pivotal studies with overall survival or progression-free survival as the primary or key secondary end point with a planned statistical comparison in the protocol. Sixty-three studies on anti-programmed cell death protein-1 or anti-programmed death/ligand 1 monoclonal antibodies used as monotherapy or in combination with other agents in 35 902 patients across multiple solid tumor types were identified. Data Extraction and Synthesis Statistical comparisons (n = 150) were made between the 3 tests using the analysis populations as defined in the original protocol of each trial. Main Outcomes and Measures Nominal significance based on a 2-sided .05-level test was used to evaluate concordance. Case studies featuring different types of nonproportional hazards were used to discuss more robust ways of characterizing treatment benefit instead of sole reliance on hazard ratios. Results In this systematic review and meta-analysis of 63 studies including 35 902 patients, between the log-rank and MaxCombo, 135 of 150 comparisons (90%) were concordant; MaxCombo achieved nominal significance in 15 of 15 discordant cases, while log-rank did not. Several cases appeared to have clinically meaningful benefits that would not have been detected using log-rank. Between the log-rank and dRMST tests, 137 of 150 comparisons (91%) were concordant; log-rank was nominally significant in 5 of 13 cases, while dRMST was significant in 8 of 13. Among all 3 tests, 127 comparisons (85%) were concordant. Conclusions and Relevance The findings of this review show that MaxCombo may provide a pragmatic alternative to log-rank when departure from proportional hazards is anticipated. Both tests resulted in the same statistical decision in most comparisons. Discordant studies had modest to meaningful improvements in treatment effect. The dRMST test provided no added sensitivity for detecting treatment differences over log-rank.
Collapse
Affiliation(s)
| | - Jiabu Ye
- Merck & Co, Inc, Kenilworth, New Jersey
| | | | | | | | - Susan Halabi
- Duke Cancer Institute, Duke University, Durham, North Carolina.,Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina
| | - Richard J Chappell
- Department of Statistics, University of Wisconsin Madison.,Department of Biostatistics and Medical Informatics, University of Wisconsin Madison
| |
Collapse
|
36
|
Smith A, Roy A, Karapetis CS, Broadbridge V, Price T. Immunotherapy use in oesophagogastric cancers-a review of the literature. Br J Cancer 2022; 127:21-29. [PMID: 35260808 PMCID: PMC9276752 DOI: 10.1038/s41416-022-01751-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 12/08/2021] [Accepted: 02/09/2022] [Indexed: 11/10/2022] Open
Abstract
Cancers of the upper gastrointestinal tract are a leading cause of cancer-related death world-wide and historically have a poor prognosis. The incidence and histology of these cancers have varied temporally and geographically over the last three decades, with an emerging understanding of the differences in the molecular and genetic profiles across different subgroups. Management of oesophagogastric cancers is by a multidisciplinary team with utilisation of surgery, radiotherapy and systemic treatments in combinations where appropriate. Immune checkpoint inhibition (ICI) has drastically changed the treatment landscape of multiple solid malignancies in the last 5 years. In oesophagogastric cancer, clinical trials have only recently shown activity that is often associated with the molecular characteristics of these tumours, in particular PD-L1 scores or microsatellite instability (MSI-H). This review looks to present the pivotal trials in this space, discuss the complexities between trials that may explain the disparate results and assess the benefit ICI offers in the treatment landscape at present.
Collapse
Affiliation(s)
- Annabel Smith
- The Queen Elizabeth Hospital, Woodville south, SA, Australia.
| | - Amitesh Roy
- Flinders Centre For Innovation in Cancer, Flinders Medical Centre, Bedford Park, SA, Australia
- Flinders University, Bedford Park, SA, Australia
| | - Christos S Karapetis
- Flinders Centre For Innovation in Cancer, Flinders Medical Centre, Bedford Park, SA, Australia
- Flinders University, Bedford Park, SA, Australia
| | - Vy Broadbridge
- The Queen Elizabeth Hospital, Woodville south, SA, Australia
| | - Timothy Price
- The Queen Elizabeth Hospital, Woodville south, SA, Australia.
- University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
37
|
Wei J, Lu X, Liu Q, Fu Y, Liu S, Li L, Liu F, Fan X, Yang J, Yang Y, Zhao Y, Guan W, Liu B. Efficacy and Safety of Sintilimab in Combination with Concurrent Chemoradiotherapy for Locally Advanced Gastric or Gastroesophageal Junction (GEJ) Adenocarcinoma (SHARED): Study Protocol of a Prospective, Multi-Center, Single-Arm Phase 2 Trial. Cancer Manag Res 2022; 14:2007-2015. [PMID: 35747712 PMCID: PMC9211075 DOI: 10.2147/cmar.s355687] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 06/01/2022] [Indexed: 12/23/2022] Open
Abstract
Purpose Concurrent chemoradiotherapy (cCRT) is the mainstay therapy of locally advanced gastric (G) and gastroesophageal junction (GEJ) cancers with a poor prognosis. Programmed cell death receptor-1 (PD-1) inhibitor has been approved and recommended to treat ≥3 line G/GEJ patients. A significant clinical benefit of PD-1 inhibitors in addition to cCRT has been observed in locally advanced lung cancer. Sintilimab, a humanized IgG4 monoclonal antibody with high affinity and specificity for PD-1, has shown promising efficacy with an overall response rate of 85% in combination with chemotherapy in gastric cancer in a phase Ib study (NCT02937116). Patients and Methods SHARED is a prospective, multicentre, single-arm Phase II trial in China, exploring the efficacy of sintilimab in combination with cCRT in locally advanced G/GEJ adenocarcinoma. According to a Simon optimal two-stage clinical design, 34 patients will be enrolled. All the eligible patients will receive one cycle of induction chemotherapy (S-1 plus nab-PTX) combined with sintilimab, followed by cCRT (radiotherapy plus nab-PTX) combined with sintilimab. Prior to the surgery, patients will receive another cycle of chemotherapy (S-1 plus nab-PTX) combined with sintilimab. In the adjuvant setting, all participants will be treated with 3 cycles of chemotherapy (S-1 plus nab-PTX) combined with sintilimab. The primary endpoint is the rate of pathological complete response (pCR). The secondary endpoints include disease-free survival (DFS), major pathological response (MPR), R0 resection rate, surgical AEs, overall survival (OS), event-free survival (EFS), and safety profile. Moreover, the prognostic value of tumor biomarkers and immune biomarkers will be explored. Conclusion SHARED is designed to primally evaluate the efficacy and safety of sintilimab in combination with cCRT in locally advanced G/GEJ cancers and to prospectively validate the prognostic value of tumor biomarkers and immune biomarkers.
Collapse
Affiliation(s)
- Jia Wei
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, People's Republic of China
| | - Xiaofeng Lu
- Department of General Surgery, Drum Tower Hospital, Nanjing, 210008, People's Republic of China
| | - Qin Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, People's Republic of China
| | - Yao Fu
- Department of Pathology, Drum Tower Hospital, Nanjing, 210008, People's Republic of China
| | - Song Liu
- Department of Radiology, Drum Tower Hospital, Nanjing, 210008, People's Republic of China
| | - Lin Li
- Department of Pathology, Drum Tower Hospital, Nanjing, 210008, People's Republic of China
| | - Fangcen Liu
- Department of Pathology, Drum Tower Hospital, Nanjing, 210008, People's Republic of China
| | - Xiangshan Fan
- Department of Pathology, Drum Tower Hospital, Nanjing, 210008, People's Republic of China
| | - Ju Yang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, People's Republic of China
| | - Yang Yang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, People's Republic of China
| | - Yang Zhao
- Department of Biostatistics, Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Wenxian Guan
- Department of General Surgery, Drum Tower Hospital, Nanjing, 210008, People's Republic of China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, People's Republic of China
| |
Collapse
|
38
|
Wang J, He Y, Zhang B, Lv H, Nie C, Chen B, Xu W, Zhao J, Cheng X, Li Q, Tu S, Chen X. The Efficacy and Safety of Sintilimab Combined With Nab-Paclitaxel as a Second-Line Treatment for Advanced or Metastatic Gastric Cancer and Gastroesophageal Junction Cancer. Front Oncol 2022; 12:924149. [PMID: 35719979 PMCID: PMC9198424 DOI: 10.3389/fonc.2022.924149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/02/2022] [Indexed: 12/19/2022] Open
Abstract
Background Unresectable advanced or recurrent gastric cancer patients have a poor prognosis. PD-1 monotherapy regimen and PD-1 combined chemotherapy regimen have become the standard third- and first-line treatment for advanced gastric cancer, respectively. However, the status of immune checkpoint inhibitors in the second-line treatment for advanced gastric cancer has not been established. The combination of chemotherapy and anti-PD-1 antibody has been demonstrated to have a synergistic effect. In this study, we aimed to evaluate the efficacy and safety of sintilimab combined with nab-paclitaxel in the second-line treatment for advanced gastric cancer (GC)/gastroesophageal junction (GEJ) cancer patients. Patients and Methods We retrospectively analyzed patients with advanced GC/GEJ cancer that progressed after first-line systemic therapies with sintilimab combined with nab-paclitaxel from April 1, 2019 to December 31, 2021. The primary endpoint was progression-free survival (PFS). The secondary endpoints included objective response rate (ORR), disease control rate (DCR), and safety. Results Thirty-nine patients were enrolled and eligible for response assessment. Complete response (CR) was not observed, 15 patients achieved partial response (PR), 16 patients had stable disease (SD) and 9 patients had progressive disease (PD). The ORR and DCR were 15 (38.5%) and 31 (79.5%), respectively. Median PFS was 5.4 months (95%CI: 3.072-7.728). PFSs between different subgroups were analyzed. The results showed that gender, age, Human epidermal growth factor receptors 2 (HER2) status, PD-L1 expression, primary tumor site and chemotherapy cycles had no significant effect on PFS. Most of the adverse events (AEs) were of grade 1-2 and manageable. The common treatment-related adverse events of grade 3 or 4 included anemia (12.8%), neutropenia (12.8%), leukopenia (10.3%), hand-foot syndrome (7.7%), thrombocytopenia (7.7%). The potential immune-related adverse events (irAEs) were grade 1 pneumonia (1 pts [2.6%]) and grade 4 hepatitis (1 pts [2.6%]). There were no treatment-related deaths. Conclusion These results indicate that sintilimab combined with nab-paclitaxel exhibits good anti-tumor activity and an acceptable safety profile as a second-line treatment for advanced or metastatic gastric cancer. These results warrant further investigation and evaluation to identify patients who can benefit more from the combined treatment strategy.
Collapse
Affiliation(s)
- Jianzheng Wang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou, University and Henan Cancer Hospital, Zhengzhou, China
| | - Yunduan He
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou, University and Henan Cancer Hospital, Zhengzhou, China
| | - Baiwen Zhang
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Huifang Lv
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou, University and Henan Cancer Hospital, Zhengzhou, China
| | - Caiyun Nie
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou, University and Henan Cancer Hospital, Zhengzhou, China
| | - Beibei Chen
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou, University and Henan Cancer Hospital, Zhengzhou, China
| | - Weifeng Xu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou, University and Henan Cancer Hospital, Zhengzhou, China
| | - Jing Zhao
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou, University and Henan Cancer Hospital, Zhengzhou, China
| | - Xiaojiao Cheng
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qingli Li
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shuiping Tu
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiaobing Chen
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou, University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
39
|
Liu M, Luo C, Xie ZZ, Li X. Treatment of gastric hepatoid adenocarcinoma with pembrolizumab and bevacizumab combination chemotherapy: A case report. World J Clin Cases 2022; 10:5420-5427. [PMID: 35812689 PMCID: PMC9210915 DOI: 10.12998/wjcc.v10.i16.5420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/19/2021] [Accepted: 04/22/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric hepatoid adenocarcinoma (GHA) is a rare and aggressive cancer that is characterized by foci with features of both hepatocellular differentiation and adenomatous differentiation. However, there is currently no standard treatment for this disease, which has a poor prognosis.
CASE SUMMARY A 72-year-old male with a body mass index of 20.9 was diagnosed with GHA with perigastric lymph node and liver metastasis. He underwent first-line chemotherapy but that failed. Pembrolizumab and bevacizumab with chemotherapy were used in the second-line treatment. The progression-free survival and overall survival were 14 mo and 16 mo, respectively, after treatment. In addition, the main adverse reaction was tolerable. The patient did not die of tumor progression.
CONCLUSION The combination of pembrolizumab and bevacizumab with chemotherapy is an effective and safe regimen for GHA and may be recommended as a new choice for GHA treatment. Further studies should evaluate this treatment in a larger cohort or a randomized controlled trial.
Collapse
Affiliation(s)
- Mei Liu
- Department of Medical Oncology, Haikou People's Hospital, Haikou 570100, Hainan Province, China
| | - Cheng Luo
- Department of Gynecology, Hainan Hospital of Chinese People’s Liberation Army General Hospital, Sanya 572013, Hainan Province, China
| | - Zong-Zhou Xie
- Department of Medical Oncology, Haikou People's Hospital, Haikou 570100, Hainan Province, China
| | - Xun Li
- Department of Medical Oncology, Haikou People's Hospital, Haikou 570100, Hainan Province, China
| |
Collapse
|
40
|
Chang X, Ge X, Zhang Y, Xue X. The current management and biomarkers of immunotherapy in advanced gastric cancer. Medicine (Baltimore) 2022; 101:e29304. [PMID: 35623069 PMCID: PMC9276259 DOI: 10.1097/md.0000000000029304] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 04/27/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Gastric carcinoma (GC) is the fourth most common cause of cancer-related death worldwide. Most patients are diagnosed at later stage, because of few treatment options, the prognosis is poor. In recent years, however, Immune checkpoint inhibitors(ICIs), such as anti- programmed death-1 (PD-1), anti-PD-L1, and anti-cytotoxic T lymphocyte antigen 4, have emerged as promising therapeutic agents in GC. Here, we summary the current treatment and advances of immune checkpoint inhibitors in the advanced stage of GC. METHODS WANFANG MED ONLINE, CNKI, NCBI PUBMED and clinicaltrials.gov were used to search literature spanning from 2000 to 2021, and all literatures about "advanced gastric or gastro-oesophageal junction cancer, Immune checkpoint inhibitors, PD-1, PD-L1, Cytotoxic T lymphocyte antigen 4, immune therapy" with detailed data were included. RESULTS Nivolumab and pembrolizumab have been recommended for the third line or subsequent therapy in advanced GC. Nivolumab plus chemotherapy has been recommended for the first line treatment in advanced GC in China. Many other ICIs have been demonstrating encouraging efficacy. PD-L1, MSI-H, Epstein Barr virus, and tumor mutational burden (TMB) status maybe potential biomarkers for response to clinical outcomes for ICIs in GC. CONCLUSION ICIs have shown encouraging treatment efficacy and manageable safety profile in GC. Some biomarkers including PD-L1, MSI-H, EBV, and TMB status could evaluate the efficacy of ICIs in GC.
Collapse
|
41
|
Yuan B, Jiang C, Chen L, Wen L, Cui J, Chen M, Zhang S, Zhou L, Cai Y, Mao JH, Zou X, Hang B, Wang P. A Novel DNA Repair Gene Signature for Immune Checkpoint Inhibitor-Based Therapy in Gastric Cancer. Front Cell Dev Biol 2022; 10:893546. [PMID: 35676932 PMCID: PMC9168368 DOI: 10.3389/fcell.2022.893546] [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: 03/10/2022] [Accepted: 04/27/2022] [Indexed: 12/07/2022] Open
Abstract
Gastric cancer is a heterogeneous group of diseases with only a fraction of patients responding to immunotherapy. The relationships between tumor DNA damage response, patient immune system and immunotherapy have recently attracted attention. Accumulating evidence suggests that DNA repair landscape is a significant factor in driving response to immune checkpoint blockade (ICB) therapy. In this study, to explore new prognostic and predictive biomarkers for gastric cancer patients who are sensitive and responsive to immunotherapies, we developed a novel 15-DNA repair gene signature (DRGS) and its related scoring system and evaluated the efficiency of the DRGS in discriminating different molecular and immune characteristics and therapeutic outcomes of patients with gastric adenocarcinoma, using publicly available datasets. The results demonstrated that DRGS high score patients showed significantly better therapeutic outcomes for ICB compared to DRGS low score patients (p < 0.001). Integrated analysis of multi-omics data demonstrated that the patients with high DRGS score were characteristic of high levels of anti-tumor lymphocyte infiltration, tumor mutation burden (TMB) and PD-L1 expression, and these patients exhibited a longer overall survival, as compared to the low-score patients. Results obtained from HPA and IHC supported significant dysregulation of the genes in DRGS in gastric cancer tissues, and a positive correlation in protein expression between DRGS and PD-L1. Therefore, the DRGS scoring system may have implications in tailoring immunotherapy in gastric cancers. A preprint has previously been published (Yuan et al., 2021).
Collapse
Affiliation(s)
- Binbin Yuan
- Department of Gastroenterology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Chengfei Jiang
- Department of Gastroenterology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lingyan Chen
- Department of Gastroenterology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lihui Wen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jinlong Cui
- Berkeley-Nanjing Research Center, Nanjing, China
| | - Min Chen
- Department of Gastroenterology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Shu Zhang
- Department of Gastroenterology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lin Zhou
- Department of Gastroenterology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yimeng Cai
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Xiaoping Zou
- Department of Gastroenterology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Bo Hang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Pin Wang
- Department of Gastroenterology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
42
|
Ma M, Sun J, Liu Z, Ouyang S, Zhang Z, Zeng Z, Li J, Kang W. The Immune Microenvironment in Gastric Cancer: Prognostic Prediction. Front Oncol 2022; 12:836389. [PMID: 35574386 PMCID: PMC9096124 DOI: 10.3389/fonc.2022.836389] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
Although therapeutic methods have been developed, gastric cancer (GC) still leads to high rates of mortality and morbidity and is the fourth leading cause of cancer-associated death and the fifth most common cancer worldwide. To understand the factors associated with the prognostic prediction of GC and to discover efficient therapeutic targets, previous studies on tumour pathogenesis have mainly focused on the cancer cells themselves; in recent years, a large number of studies have shown that cancer invasion and metastasis are the results of coevolution between cancer cells and the microenvironment. It seems that studies on the tumour microenvironment could help in prognostic prediction and identify potential targets for treating GC. In this review, we mainly introduce the research progress for prognostic prediction and the immune microenvironment in GC in recent years, focusing on cancer-associated fibroblasts (CAFs), tumour-associated macrophages (TAMs), and tumour-infiltrating lymphocytes (TILs) in GC, and discuss the possibility of new therapeutic targets for GC.
Collapse
Affiliation(s)
- Mingwei Ma
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Juan Sun
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Zhen Liu
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Siwen Ouyang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Zimu Zhang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Ziyang Zeng
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Jie Li
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Weiming Kang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
43
|
Dos Santos M, Lequesne J, Leconte A, Corbinais S, Parzy A, Guilloit JM, Varatharajah S, Brachet PE, Dorbeau M, Vaur D, Weiswald LB, Poulain L, Le Gallic C, Castera-Tellier M, Galais MP, Clarisse B. Perioperative treatment in resectable gastric cancer with spartalizumab in combination with fluorouracil, leucovorin, oxaliplatin and docetaxel (FLOT): a phase II study (GASPAR). BMC Cancer 2022; 22:537. [PMID: 35549674 PMCID: PMC9097175 DOI: 10.1186/s12885-022-09623-z] [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: 04/07/2022] [Accepted: 04/29/2022] [Indexed: 12/24/2022] Open
Abstract
Background Perioperative chemotherapy and surgery are a standard of care for patients with resectable gastric or gastroesophageal junction (GEJ) adenocarcinoma. However, the prognosis remains poor for this population. The FLOT (fluorouracil, leucovorin, oxaliplatin, and docetaxel) regimen is considered as the new standard chemotherapy regimen for perioperative strategy, despite associated with a 5-year overall survival rate (OS) amounting 45% following radical surgery. Immunotherapy with antibodies that inhibit PD-1/ PD-L1 interaction has recently emerged as a new treatment option with promising and encouraging early trial results for patients with advanced or metastatic gastric or GEJ adenocarcinoma. Currently, no trials have investigated the impact of perioperative immunotherapy in combination with chemotherapy for resectable gastric or GEJ adenocarcinoma. Methods GASPAR trial is a multicenter open-label, nonrandomized, phase II trial to evaluate the efficacy and safety of Spartalizumab in combination with the FLOT regimen as perioperative treatment for resectable gastric or GEJ adenocarcinoma. The main endpoint is the proportion of patients with pathological complete regression (pCR) in the primary tumour after preoperative treatment. Systemic treatment will include a pre-operative neoadjuvant and a post-operative adjuvant treatment, during which FLOT regimen will be administered every two weeks for 4 cycles and Spartalizumab every four weeks for 2 cycles. For patients with confirmed tumor resectability on imaging assessment, surgery will be realized within 4–6 weeks after the last dose of preoperative chemotherapy. Post-operative systemic treatment will then be initiated within 4–10 weeks after surgery. Using a Simon’s two-stage design, up to 67 patients will be enrolled, including 23 in the first stage. Discussion Currently, no trials have investigated the impact of immunotherapy in combination with FLOT chemotherapy as perioperative treatment for resectable gastric or GEJ adenocarcinoma. Some studies have suggested a change in the tumor immune micro-environment following neoadjuvant chemotherapy in this setting, reinforcing the relevance to propose a phase II trial evaluating efficacy and safety of Spartalizumab in combination with perioperative chemotherapy, with the aim of improving treatment efficacy and survival outcomes. Trial registration NCT04736485, registered February, 3, 2021.
Collapse
Affiliation(s)
- Mélanie Dos Santos
- Clinical Research Department, UNICANCER, Centre François Baclesse, 3 Avenue du Général Harris, 14000, Caen, France. .,Department of Medical Oncology, UNICANCER, Centre François Baclesse, 3 Avenue du Général Harris, 14000, Caen, France.
| | - Justine Lequesne
- Clinical Research Department, UNICANCER, Centre François Baclesse, 3 Avenue du Général Harris, 14000, Caen, France
| | - Alexandra Leconte
- Clinical Research Department, UNICANCER, Centre François Baclesse, 3 Avenue du Général Harris, 14000, Caen, France
| | - Stéphane Corbinais
- Department of Medical Oncology, UNICANCER, Centre François Baclesse, 3 Avenue du Général Harris, 14000, Caen, France
| | - Aurélie Parzy
- Department of Medical Oncology, UNICANCER, Centre François Baclesse, 3 Avenue du Général Harris, 14000, Caen, France
| | - Jean-Marc Guilloit
- Department of Surgery, UNICANCERCentre François Baclesse, 14000, Caen, France
| | | | - Pierre-Emmanuel Brachet
- Clinical Research Department, UNICANCER, Centre François Baclesse, 3 Avenue du Général Harris, 14000, Caen, France.,Department of Medical Oncology, UNICANCER, Centre François Baclesse, 3 Avenue du Général Harris, 14000, Caen, France
| | - Marine Dorbeau
- Department of Pathology, UNICANCERCentre François Baclesse, 14000, Caen, France
| | - Dominique Vaur
- Department of Cancer Biology and Genetics, UNICANCERCentre François Baclesse, 14000, Caen, France
| | - Louis-Bastien Weiswald
- UNICANCER, Centre François Baclesse, 14000, Caen, France.,Normandie University, UNICAEN, ANTICIPE, ORGAPRED Platform, Caen, France
| | - Laurent Poulain
- UNICANCER, Centre François Baclesse, 14000, Caen, France.,Normandie University, UNICAEN, ANTICIPE, ORGAPRED Platform, Caen, France
| | - Corentin Le Gallic
- Clinical Research Department, UNICANCER, Centre François Baclesse, 3 Avenue du Général Harris, 14000, Caen, France
| | - Marie Castera-Tellier
- Clinical Research Department, UNICANCER, Centre François Baclesse, 3 Avenue du Général Harris, 14000, Caen, France
| | - Marie-Pierre Galais
- Department of Medical Oncology, UNICANCER, Centre François Baclesse, 3 Avenue du Général Harris, 14000, Caen, France
| | - Bénédicte Clarisse
- Clinical Research Department, UNICANCER, Centre François Baclesse, 3 Avenue du Général Harris, 14000, Caen, France
| |
Collapse
|
44
|
Zito Marino F, Amato M, Ronchi A, Panarese I, Ferraraccio F, De Vita F, Tirino G, Martinelli E, Troiani T, Facchini G, Pirozzi F, Perrotta M, Incoronato P, Addeo R, Selvaggi F, Lucido FS, Caraglia M, Savarese G, Sirica R, Casillo M, Lieto E, Auricchio A, Cardella F, Docimo L, Galizia G, Franco R. Microsatellite Status Detection in Gastrointestinal Cancers: PCR/NGS Is Mandatory in Negative/Patchy MMR Immunohistochemistry. Cancers (Basel) 2022; 14:2204. [PMID: 35565332 PMCID: PMC9102010 DOI: 10.3390/cancers14092204] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/21/2022] [Accepted: 04/24/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Microsatellite instability (MSI) is a predictive biomarker for immune checkpoint inhibitors. The main goal was to investigate the discordance between IHC and PCR/NGS for MSI testing in gastrointestinal cancers. METHODS Two series were analyzed through IHC for mismatch-repair-system proteins (MMRP) and PCR, with one series of 444 colorectal cancers (CRC) and the other of 176 gastric cancers (GC). All cases with discordant results between IHC and PCR were analyzed by NGS. IHC staining was evaluated as follows: proficient MMR (pMMR), with all MMR positive; deficient MMR (dMMR), with the loss of one heterodimer; and cases with the loss/patchy expression of one MMR (lo-paMMR). Cases with instability in at least two markers by PCR were MSI-high (MSI-H) and with instability in one marker, MSI-low (MSI-L). Cases without instability were evaluated as microsatellite-stable (MSS). RESULTS In the CRC cohort, 15 out of 444 cases were dMMR and 46 lo-paMMR. Among the 15 dMMR, 13 were MSI-H and 2 MSS. Among the 46 lo-paMMR, 13 were MSI-H and 33 were MSS. In the GC cohort, 13 out of 176 cases were dMMR and 6 cases lo-paMMR. Among the 13 dMMR, 12 were MSI-H and only 1 was MSS. All six lo-paMMR cases were MSS. All NGS results were in agreement with PCR. CONCLUSIONS In clinical practice, MMR-IHC could be used as a screening test and additional molecular analysis is mandatory exclusively in cases carrying loss/patchy MMR-IHC.
Collapse
Affiliation(s)
- Federica Zito Marino
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| | - Martina Amato
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| | - Andrea Ronchi
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| | - Iacopo Panarese
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| | - Franca Ferraraccio
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| | - Ferdinando De Vita
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.D.V.); (G.T.); (E.M.); (T.T.); (M.C.)
| | - Giuseppe Tirino
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.D.V.); (G.T.); (E.M.); (T.T.); (M.C.)
| | - Erika Martinelli
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.D.V.); (G.T.); (E.M.); (T.T.); (M.C.)
| | - Teresa Troiani
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.D.V.); (G.T.); (E.M.); (T.T.); (M.C.)
| | - Gaetano Facchini
- Medical Oncology Unit, SM delle Grazie Hospital, 80078 Pozzuoli, Naples, Italy;
| | - Felice Pirozzi
- General Surgery Unit, Santa Maria delle Grazie Hospital, 80078 Pozzuoli, Naples, Italy;
| | - Michele Perrotta
- Hepatology and Interventional Ultrasound Unit, San Giovanni di Dio Hospital, 80027 Frattamaggiore, Naples, Italy;
| | - Pasquale Incoronato
- Medical Oncology Unit, ASL Napoli 2 Nord Hospital, 80014 Giugliano, Naples, Italy;
| | - Raffaele Addeo
- Medical Oncology Unit, San Giovanni di Dio Hospital, 80027 Frattamaggiore, Naples, Italy;
| | - Francesco Selvaggi
- Department of Advanced Medical and Surgical Sciences, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (F.S.); (F.S.L.); (L.D.)
| | - Francesco Saverio Lucido
- Department of Advanced Medical and Surgical Sciences, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (F.S.); (F.S.L.); (L.D.)
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.D.V.); (G.T.); (E.M.); (T.T.); (M.C.)
| | - Giovanni Savarese
- AMES, Centro Polidiagnostico Strumentale srl, 80013 Casalnuovo, Naples, Italy; (G.S.); (R.S.); (M.C.)
| | - Roberto Sirica
- AMES, Centro Polidiagnostico Strumentale srl, 80013 Casalnuovo, Naples, Italy; (G.S.); (R.S.); (M.C.)
| | - Marika Casillo
- AMES, Centro Polidiagnostico Strumentale srl, 80013 Casalnuovo, Naples, Italy; (G.S.); (R.S.); (M.C.)
| | - Eva Lieto
- Department of Translational Medical Science, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (E.L.); (A.A.); (F.C.)
| | - Annamaria Auricchio
- Department of Translational Medical Science, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (E.L.); (A.A.); (F.C.)
| | - Francesca Cardella
- Department of Translational Medical Science, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (E.L.); (A.A.); (F.C.)
| | - Ludovico Docimo
- Department of Advanced Medical and Surgical Sciences, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (F.S.); (F.S.L.); (L.D.)
| | - Gennaro Galizia
- Department of Surgical Sciences, School of Medicine, Second University of Naples, Place Miraglia, 3th Building, West Side, 4th Floor, 80138 Naples, Italy;
| | - Renato Franco
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| |
Collapse
|
45
|
Mylod E, McKenna E, Davern M, Barr MP, Donlon NE, Bibby BAS, Bhardwaj A, Reynolds JV, Lysaght J, Maher SG, Conroy MJ. Investigating the susceptibility of treatment-resistant oesophageal tumours to natural killer cell-mediated responses. Clin Exp Med 2022; 23:411-425. [PMID: 35364779 DOI: 10.1007/s10238-022-00811-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 02/22/2022] [Indexed: 11/26/2022]
Abstract
The majority of oesophageal adenocarcinoma (OAC) patients do not respond to multimodal treatment regimens and face dismal survival rates. Natural killer (NK) cells are crucial anti-tumour immune cells, and this study investigated the susceptibility of treatment-resistant OAC cells to these potent tumour killers. Natural killer receptor (NKR) ligand expression by OE33CisP (cisplatin-sensitive) and OE33CisR (cisplatin-resistant) cells was investigated. The immunomodulatory effects of OE33CisP and OE33CisR cells on NK cell phenotype and function were assessed. Finally, the impact of chemotherapy regimens on NKR ligand shedding was examined. Our data revealed significantly less surface expression of activating ligands B7-H6, MICA/B, ULBP-3 and activating/inhibitory ligands PVRL-1 and PVRL-4 by OE33CisR cells, compared to OE33CisP cells. Co-culture with OE33CisR cells reduced the frequencies of NKp30+ and NKp46+ NK cells and increased frequencies of TIGIT+, FasL+ and TRAIL+ NK cells. Frequencies of IFN-γ-producing NK cells increased while frequencies of TIM-3+ NK cells decreased after culture with OE33CisP and OE33CisR cells. Frequencies of circulating NKp30+ NK cells were significantly lower in OAC patients with the poorest treatment response and in patients who received FLOT chemotherapy, while B7-H6 shedding by OAC tumour cells was induced by FLOT. Overall, OE33CisR cells express less activating NKR ligands than OE33CisP cells and have differential effects on NKR expression by NK cells. However, neither cell line significantly dampened NK cell cytokine production, death receptor expression or degranulation. In addition, our data indicate that FLOT chemotherapy may promote B7-H6 shedding and immune evasion with detrimental consequences in OAC patients.
Collapse
Affiliation(s)
- Eimear Mylod
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - Ellen McKenna
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - Maria Davern
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - Martin P Barr
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - Noel E Donlon
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - Becky A S Bibby
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, M20 4BX, UK
| | - Anshul Bhardwaj
- Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - John V Reynolds
- Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
- National Oesophageal and Gastric Centre, St. James's Hospital, Dublin, Ireland
| | - Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - Stephen G Maher
- Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - Melissa J Conroy
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland.
- Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland.
| |
Collapse
|
46
|
Advances in the curative management of oesophageal cancer. Br J Cancer 2022; 126:706-717. [PMID: 34675397 PMCID: PMC8528946 DOI: 10.1038/s41416-021-01485-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/01/2021] [Accepted: 06/30/2021] [Indexed: 12/24/2022] Open
Abstract
The incidence of oesophageal cancer, in particular adenocarcinoma, has markedly increased over the last four decades with adenocarcinoma becoming the dominant subtype in the West, and mortality rates are high. Nevertheless, overall survival of patients with oesophageal cancer has doubled in the past 20 years, with earlier diagnosis and improved treatments benefiting those patients who can be treated with curative intent. Advances in endotherapy, surgical approaches, and multimodal and other combination therapies have been reported. New vistas have emerged in targeted therapies and immunotherapy, informed by new knowledge in genomics and molecular biology, which present opportunities for personalised cancer therapy and novel clinical trials. This review focuses exclusively on the curative intent treatment pathway, and highlights emerging advances.
Collapse
|
47
|
Jiang H, Yu X, Li N, Kong M, Ma Z, Zhou D, Wang W, Wang H, Wang H, He K, Li Z, Lu Y, Zhang J, Zhao K, Zhang Y, Xu N, Li Z, Liu Y, Wang Y, Wang Y, Teng L. Efficacy and safety of neoadjuvant sintilimab, oxaliplatin and capecitabine in patients with locally advanced, resectable gastric or gastroesophageal junction adenocarcinoma: early results of a phase 2 study. J Immunother Cancer 2022; 10:jitc-2021-003635. [PMID: 35296556 PMCID: PMC8928365 DOI: 10.1136/jitc-2021-003635] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2022] [Indexed: 02/03/2023] Open
Abstract
Immune checkpoint inhibitors have greatly improved the prognoses of diverse advanced malignancies, including gastric and gastroesophageal junction (G/GEJ) cancer. However, the role of anti-programmed cell death protein-1 treatment in the neoadjuvant setting remains unclear. This phase 2 study aimed to evaluate sintilimab plus CapeOx as a neoadjuvant regimen in patients with advanced resectable G/GEJ adenocarcinoma. Eligible patients with resectable G/GEJ adenocarcinoma stage cT3-4NanyM0 were enrolled. Patients received neoadjuvant treatment with sintilimab (3 mg/kg for cases <60 kg or 200 mg for those ≥60 kg on day 1) plus CapeOx (oxaliplatin at 130 mg/m2 on D1 and capecitabine at 1000 mg/m2 two times per day on D1-D14) every 21 days, for three cycles before surgical resection, followed by adjuvant treatment with three cycles of CapeOx with the same dosages after surgical resection. The primary endpoint was pathological complete response (pCR) rate. Secondary endpoints included objective response rate, tumor regression grade per Becker criteria, survival and safety. As of July 30, 2020, 36 patients were enrolled. Totally 7 (19.4%) patients had GEJ cancer, and 34 (94.4%) patients were clinical stage III cases. A total of 35 (97.2%) patients completed three cycles of neoadjuvant treatment, and 1 patients received two cycles due to adverse events. All patients underwent surgery and the R0 resection rate was 97.2%. In this study, pCR and major pathological response were achieved in 7 (19.4%, 95% CI: 8.8% to 35.7%; 90% CI: 10.7% to 33.1%) and 17 (47.2%, 95% CI: 31.6% to 64.3%) patients, respectively. Thirty-one patients received adjuvant treatment. By December 20, 2021, three patients died after disease relapse, and two patients were alive with relapse. Median disease-free survival (DFS) and overall survival (OS) were not reached. The 1-year DFS and OS rates were 90.3% (95% CI: 80.4% to 100.0%) and 94.1% (95% CI: 86.5% to 100.0%), respectively. The most common (>1 patient) grade 3 treatment-related adverse events during neoadjuvant treatment were anemia and neutropenia (n=5 each, 13.9%). No serious adverse events (AEs) or grade 4-5 AEs were observed. Sintilimab plus oxaliplatin/capecitabine showed promising efficacy with encouraging pCR rate and good safety profile in the neoadjuvant setting. This combination regimen might present a new option for patients with locally advanced, resectable G/GEJ adenocarcinoma. Trial registration; NCT04065282.
Collapse
Affiliation(s)
- Haiping Jiang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiongfei Yu
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ning Li
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mei Kong
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhimin Ma
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Donghui Zhou
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weibin Wang
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haohao Wang
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haiyong Wang
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kuifeng He
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhongqi Li
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yimin Lu
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Zhang
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kui Zhao
- Department of Nuclear Medicine, PET Centre, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yafei Zhang
- Department of Nuclear Medicine, PET Centre, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziran Li
- Department of Medical Science and Strategy Oncology, Innovent Biologics, Inc, Suzhou, China
| | - Ying Liu
- Department of Medical Science and Strategy Oncology, Innovent Biologics, Inc, Suzhou, China
| | - Yan Wang
- Department of Medical Science and Strategy Oncology, Innovent Biologics, Inc, Suzhou, China
| | - Yisen Wang
- Department of Translational Medicine, Innovent Biologics, Inc, Suzhou, China
| | - Lisong Teng
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
48
|
Ignatova EO, Kozlov E, Ivanov M, Mileyko V, Menshikova S, Sun H, Fedyanin M, Tryakin A, Stilidi I. Clinical significance of molecular subtypes of gastrointestinal tract adenocarcinoma. World J Gastrointest Oncol 2022; 14:628-645. [PMID: 35321271 PMCID: PMC8919013 DOI: 10.4251/wjgo.v14.i3.628] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/04/2021] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
Adenocarcinomas of the gastrointestinal tract (esophagus, stomach, and colon) represent a heterogeneous group of diseases with distinct etiology, clinical features, treatment approaches, and prognosis. Studies are ongoing to isolate molecular genetic subtypes, perform complete biological characterization of the tumor, determine prognostic groups, and find predictive markers to the effectiveness of therapy. Separate molecular genetic classifications were created for esophageal adenocarcinoma [The Cancer Genome Atlas (TCGA)], stomach cancer (TCGA, Asian Cancer Research Group), and colon cancer (Colorectal Cancer Subtyping Consortium). In 2018, isolation of TCGA molecular genetic subtypes for adenocarcinomas of the gastrointestinal tract (esophagus, stomach, and colon) highlighted the need for further studies and clinical validation of subtyping of gastrointestinal adenocarcinomas. However, this approach has limitations. The aim of our work was to critically analyze integration of molecular genetic subtyping of gastrointestinal adenocarcinomas in clinical practice.
Collapse
Affiliation(s)
- Ekaterina Olegovna Ignatova
- Department of Second Chemotherapy, Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow 115478, Moscow, Russia
- Department of Oncogenetics, Research Centre for Medical Genetics Research Centre for Medical Genetics, Moscow 115522, Moscow, Russia
| | | | - Maxim Ivanov
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Moscow 141700, Moscow, Russia
| | | | - Sofia Menshikova
- Department of Anticancer Drug Treatment, AO K31 City, Moscow 121552, Moscow, Russia
| | - Henian Sun
- Pirogov Russian National Research Medical University (Pirogov Medical University), Moscow 117997, Moscow, Russia
| | - Mikhail Fedyanin
- Department of Second Chemotherapy, Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow 115478, Moscow, Russia
| | - Alexey Tryakin
- Department of Second Chemotherapy, Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow 115478, Moscow, Russia
| | - Ivan Stilidi
- Department of Second Chemotherapy, Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow 115478, Moscow, Russia
| |
Collapse
|
49
|
Xing X, Shi J, Jia Y, Dou Y, Li Z, Dong B, Guo T, Cheng X, Li X, Du H, Hu Y, Jia S, Zhang J, Li Z, Ji J. Effect of neoadjuvant chemotherapy on the immune microenvironment in gastric cancer as determined by multiplex immunofluorescence and T cell receptor repertoire analysis. J Immunother Cancer 2022; 10:e003984. [PMID: 35361730 PMCID: PMC8971786 DOI: 10.1136/jitc-2021-003984] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The combination of immune checkpoint blockade and chemotherapy has revolutionized the treatment of advanced gastric cancer (GC). It is crucial to unravel chemotherapy-induced tumor microenvironment (TME) modulation and identify which immunotherapy would improve antitumor effect. METHODS In this study, tumor-associated immune cells (TAICs) infiltration in residual tumor after neoadjuvant chemotherapy (NAC) together with 1075 cases of treatment-naïve GC patients was analyzed first. Then we performed multiplex fluorescence staining of a panel of immune markers (CD3, CD4, CD8, FOXP3 and PDL1) and T cell receptor β-chain sequencing to phenotype and enumerate T cell subpopulations and clonal expansion in paired GC samples (prechemotherapy and postchemotherapy) from another cohort of 30 cases of stage II/III GC patients. RESULTS Infiltration of CD68+ macrophages in residual tumors after NAC was significantly decreased compared with treatment-naïve GC patients, while no significant difference observed with respect to other immune markers. In residual tumors, post-NAC CD8 +T cells and CD68+ macrophages levels were significantly associated with chemotherapy response. Post-NAC CD8+ T cell levels remained as an independent predictor for favorable prognosis. Furthermore, when comparing the paired samples before and after NAC from 30 cases of stage II/III GC patients, we found FOXP3+ regulatory T cells proportion significantly decreased after chemotherapy. Pre-NAC FOXP3+ T reg cells level was much richer in the response group and decreased more significantly in the stromal compartment. CD8+ cytotoxic T lymphocytes levels were elevated after chemotherapy, which was more significant in the group treated with XELOX regimen and in patients with better response, consistent with the TCR diversity elevation. CONCLUSIONS These findings have deepened our understanding of the immune modulating effect of chemotherapy and suggest that the immune profile of specimens after standard chemotherapy should be considered for the personalized immunotherapy to ultimately improve clinical outcome in patients with GC.
Collapse
Affiliation(s)
- Xiaofang Xing
- Department of Gastrointestinal Translational Research, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing, China
| | - Jinyao Shi
- Department of Gastrointestinal Translational Research, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing, China
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yongning Jia
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yunsheng Dou
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Zhongwu Li
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Bin Dong
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Ting Guo
- Department of Gastrointestinal Translational Research, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing, China
| | - Xiaojing Cheng
- Department of Gastrointestinal Translational Research, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing, China
| | - Xiaomei Li
- Department of Gastrointestinal Translational Research, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing, China
| | - Hong Du
- Department of Gastrointestinal Translational Research, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing, China
| | - Ying Hu
- Biobank, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Shuqin Jia
- Department of Molecular Diagnosis, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jian Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Ziyu Li
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiafu Ji
- Department of Gastrointestinal Translational Research, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing, China
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
50
|
Konopka K, Frączek P, Lubaś M, Micek A, Kwinta Ł, Streb J, Potocki P, Wysocki PJ. Reduction of Cancer-Induced Thrombocytosis as a Biomarker of Improved Outcomes in Advanced Gastric Cancer. J Clin Med 2022; 11:jcm11051213. [PMID: 35268305 PMCID: PMC8911022 DOI: 10.3390/jcm11051213] [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: 02/08/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Interplay between non-specific inflammatory reaction and tumor microenvironment in gastric cancer (GC) can be measured indirectly by assessing fluctuations in concentration of platelets. Cytotoxic chemotherapy affects these morphotic elements directly by inducing myelosuppression. It was hypothesized that chemotherapy not only directly affects malignant cells, but also through immunomodulation related to myelosuppression. METHODS Metastatic GC patients (N: 155) treated with chemotherapy +/- trastuzumab were enrolled in this retrospective study. Platelet pretreatment concentration (PLT-count) and the deepest level of platelet reduction, as well as other inflammatory and general confounders were collected in the first 12 weeks of treatment (PLT-red). Martingale residuals were used to visualize the relationship between PLT-count, PLT-red, and overall survival (OS). Multiple multivariate Cox regression models were built to assess the impact of platelet reduction on OS and progression-free survival (PFS). RESULTS Reduction of PLT (PLT-red) to 60% of baseline concentration was associated with improved survival rates (HR = 0.60, p = 0.026 for OS and HR 0.56, p = 0.015 for PFS). Cross-classification into four groups based on PLT-count (high vs low) and PLT-red (high vs low) showed significantly worse survival rates in both high PLT-count (HR = 3.60, p = 0.007 for OS and HR = 2.97, p = 0.024 for PFS) and low PLT-count (HR = 1.75, p = 0.035 for OS and HR = 1.80, p = 0.028 for PFS) patients with insufficient platelets reduction. CONCLUSION Thrombocytosis reduction represents a novel, clinically important, prognostic factor for OS and PFS in patients with stage IV GC.
Collapse
Affiliation(s)
- Kamil Konopka
- Department of Oncology, Jagiellonian University Medical College, 31-007 Cracow, Poland
- Correspondence:
| | - Paulina Frączek
- Department of Medical Oncology, University Hospital in Cracow, 30-688 Cracow, Poland
| | - Maciej Lubaś
- Department of Medical Oncology, University Hospital in Cracow, 30-688 Cracow, Poland
| | - Agnieszka Micek
- Department of Nursing Management and Epidemiology Nursing, Jagiellonian University Medical College, 31-007 Cracow, Poland
| | - Łukasz Kwinta
- Department of Oncology, Jagiellonian University Medical College, 31-007 Cracow, Poland
| | - Joanna Streb
- Department of Oncology, Jagiellonian University Medical College, 31-007 Cracow, Poland
| | - Paweł Potocki
- Department of Oncology, Jagiellonian University Medical College, 31-007 Cracow, Poland
| | - Piotr J. Wysocki
- Department of Oncology, Jagiellonian University Medical College, 31-007 Cracow, Poland
| |
Collapse
|