1
|
Zhang P, Zhang X, Zhu N, Zhuang F, Zhou D, Wang P. Clinical Efficacy, Survival, and Adverse Reaction Evaluation of Immune Checkpoint Inhibitor in Advanced Gastric Cancer: A Systematic Review and Meta-Analysis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:6998090. [PMID: 36050997 PMCID: PMC9427293 DOI: 10.1155/2022/6998090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/15/2022] [Accepted: 07/23/2022] [Indexed: 11/20/2022]
Abstract
Objective To systematically assess the clinical effect and survival time of immune checkpoint inhibitor (ICIs) in advanced gastric cancer (GC) and adverse reactions to provide evidence-based medicine for its enhancement and adoption. Methods PubMed, EMBASE, ScienceDirect, Cochrane Library, China Knowledge Network database (CNKI), China VIP database, Wanfang database, and China Biomedical Literature Database (CBM) online database were searched for randomized controlled trials (RCT) of immuno-checkpoint inhibitors in advanced GC therapy. Retrieval time was limited to the period from the date the database was established to present. Separately, two researchers gathered the data. Statistical software RevMan5.4 was used to estimate bias risk according to the Cochrane Handbook 5.3 standard. Results The computer database retrieved 1723 articles, and 465 articles were eliminated when repeated studies were removed. After screening the titles and abstracts of 287 articles, 124 articles were contained after eliminating irrelevant studies, reviews, case reports, and no control literature. After carefully reading 108 studies with insufficient data and no major outcome markers, 6 RCTs were eventually contained. 4 articles compared the levels of carcinoembryonic antigen (CEA) and carbohydrate antigen 199 (CA199) after treatment. The result indicated that the levels of serum CEA and CA199 in the study group were notably lower, and the difference was statistically significant (P < 0.05). The immune function indexes after treatment were compared, suggesting that the improvement of immune function indexes in the study group was notably better, and the difference was statistically significant (P < 0.05). Three clinical trials reported the median progression-free survival (PFS). The PFS of the study group was notably longer after treatment, and the difference was statistically significant (P < 0.05). The occurrence of adverse reactions after treatment was analyzed by meat, and all the literatures were analyzed. No notable differences were observed in the incidence of adverse reactions. Conclusion ICIs associated with chemotherapy is effective when treating GC, which can effectively promote the disease control rate of patients, enhance immune function, reduce the level of tumor markers, and prolong survival time. The safety is controllable, which is worth popularizing in clinical practice. However, more studies and follow-up with higher methodological quality and longer intervention time are needed to further verify it.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Medical Oncology, Yantaishan Hospital, Yantai 264003, China
| | - Xiaomeng Zhang
- Public Health Division, Yantaishan Hospital, Yantai 264003, China
| | - Na Zhu
- Department of Medical Oncology, Yantaishan Hospital, Yantai 264003, China
| | - Feifei Zhuang
- Department of Medical Oncology, Yantaishan Hospital, Yantai 264003, China
| | - Dongmei Zhou
- Department of Medical Oncology, Yantaishan Hospital, Yantai 264003, China
| | - Ping Wang
- Department of Medical Oncology, Yantaishan Hospital, Yantai 264003, China
| |
Collapse
|
2
|
Pozzi S, Scomparin A, Israeli Dangoor S, Rodriguez Ajamil D, Ofek P, Neufeld L, Krivitsky A, Vaskovich-Koubi D, Kleiner R, Dey P, Koshrovski-Michael S, Reisman N, Satchi-Fainaro R. Meet me halfway: Are in vitro 3D cancer models on the way to replace in vivo models for nanomedicine development? Adv Drug Deliv Rev 2021; 175:113760. [PMID: 33838208 DOI: 10.1016/j.addr.2021.04.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/24/2021] [Accepted: 04/01/2021] [Indexed: 12/12/2022]
Abstract
The complexity and diversity of the biochemical processes that occur during tumorigenesis and metastasis are frequently over-simplified in the traditional in vitro cell cultures. Two-dimensional cultures limit researchers' experimental observations and frequently give rise to misleading and contradictory results. Therefore, in order to overcome the limitations of in vitro studies and bridge the translational gap to in vivo applications, 3D models of cancer were developed in the last decades. The three dimensions of the tumor, including its cellular and extracellular microenvironment, are recreated by combining co-cultures of cancer and stromal cells in 3D hydrogel-based growth factors-inclusive scaffolds. More complex 3D cultures, containing functional blood vasculature, can integrate in the system external stimuli (e.g. oxygen and nutrient deprivation, cytokines, growth factors) along with drugs, or other therapeutic compounds. In this scenario, cell signaling pathways, metastatic cascade steps, cell differentiation and self-renewal, tumor-microenvironment interactions, and precision and personalized medicine, are among the wide range of biological applications that can be studied. Here, we discuss a broad variety of strategies exploited by scientists to create in vitro 3D cancer models that resemble as much as possible the biology and patho-physiology of in vivo tumors and predict faithfully the treatment outcome.
Collapse
Affiliation(s)
- Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anna Scomparin
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Turin, Italy
| | - Sahar Israeli Dangoor
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Daniel Rodriguez Ajamil
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Paula Ofek
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Lena Neufeld
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Adva Krivitsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Daniella Vaskovich-Koubi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ron Kleiner
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Pradip Dey
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shani Koshrovski-Michael
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Noa Reisman
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
3
|
de Fátima Aquino Moreira-Nunes C, de Souza Almeida Titan Martins CN, Feio D, Lima IK, Lamarão LM, de Souza CRT, Costa IB, da Silva Maués JH, Soares PC, de Assumpção PP, Burbano RMR. PD-L1 Expression Associated with Epstein-Barr Virus Status and Patients' Survival in a Large Cohort of Gastric Cancer Patients in Northern Brazil. Cancers (Basel) 2021; 13:3107. [PMID: 34206307 PMCID: PMC8268941 DOI: 10.3390/cancers13133107] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022] Open
Abstract
Gastric cancer (GC) is a worldwide health problem, making it one of the most common types of cancer, in fifth place of all tumor types, and the third highest cause of cancer deaths in the world. There is a subgroup of GC that consists of tumors infected with the Epstein-Barr virus (EBV) and is characterized mainly by the overexpression of programmed cell death protein-ligand-1 (PD-L1). In the present study, we present histopathological and survival data of a thousand GC patients, associated with EBV status and PD-L1 expression. Of the thousand tumors analyzed, 190 were EBV-positive and the vast majority (86.8%) had a high relative expression of mRNA and PD-L1 protein (p < 0.0001) in relation to non-neoplastic control. On the other hand, in EBV-negative samples, the majority had a low PD-L1 expression of RNA and protein (p < 0.0001). In the Kaplan-Meier analysis, the probability of survival and increased overall survival of EBV-positive GC patients was impacted by the PD-L1 overexpression (p < 0.0001 and p = 0.004, respectively). However, the PD-L1 low expression was correlated with low overall survival in those patients. Patients with GC positive for EBV, presenting PD-L1 overexpression can benefit from immunotherapy treatments and performing the quantification of PD-L1 in gastric neoplasms should be adopted as routine.
Collapse
Affiliation(s)
- Caroline de Fátima Aquino Moreira-Nunes
- Laboratory of Molecular Biology, Department of Clinical Medicine, Ophir Loyola Hospital, Belém, 66063-240 PA, Brazil; (C.N.d.S.A.T.M.); (D.F.); (I.K.L.); (P.C.S.)
- Laboratory of Pharmacogenetics, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, 60430-275 CE, Brazil
| | | | - Danielle Feio
- Laboratory of Molecular Biology, Department of Clinical Medicine, Ophir Loyola Hospital, Belém, 66063-240 PA, Brazil; (C.N.d.S.A.T.M.); (D.F.); (I.K.L.); (P.C.S.)
| | - Isamu Komatsu Lima
- Laboratory of Molecular Biology, Department of Clinical Medicine, Ophir Loyola Hospital, Belém, 66063-240 PA, Brazil; (C.N.d.S.A.T.M.); (D.F.); (I.K.L.); (P.C.S.)
| | - Leticia Martins Lamarão
- Foundation Center for Hemotherapy and Hematology of Pará (HEMOPA), Department of Sorology, Belém, 66033-000 PA, Brazil;
| | | | - Igor Brasil Costa
- Department of Virology, Evandro Chagas Institute, Ananindeua, 67030-000 PA, Brazil;
| | - Jersey Heitor da Silva Maués
- Hematology and Transfusion Medicine Center, Laboratory of Molecular and Cell Biology, Department of Medicine, University of Campinas, Campinas, 13083-970 SP, Brazil;
| | - Paulo Cardoso Soares
- Laboratory of Molecular Biology, Department of Clinical Medicine, Ophir Loyola Hospital, Belém, 66063-240 PA, Brazil; (C.N.d.S.A.T.M.); (D.F.); (I.K.L.); (P.C.S.)
| | - Paulo Pimentel de Assumpção
- Oncology Research Center, Department of Biological Sciences, Federal University of Pará, Belém, 66073-005 PA, Brazil;
| | - Rommel Mário Rodríguez Burbano
- Laboratory of Molecular Biology, Department of Clinical Medicine, Ophir Loyola Hospital, Belém, 66063-240 PA, Brazil; (C.N.d.S.A.T.M.); (D.F.); (I.K.L.); (P.C.S.)
- Oncology Research Center, Department of Biological Sciences, Federal University of Pará, Belém, 66073-005 PA, Brazil;
| |
Collapse
|
4
|
Comparison of PD-L1 immunohistochemical assays in advanced gastric adenocarcinomas using endoscopic biopsy and paired resected specimens. Pathology 2021; 53:586-594. [PMID: 33546812 DOI: 10.1016/j.pathol.2020.10.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/24/2020] [Accepted: 10/05/2020] [Indexed: 11/21/2022]
Abstract
Immunohistochemical (IHC) assays for programmed death ligand 1 (PD-L1) expression are crucial for guiding immune checkpoint inhibitor therapies in advanced gastric adenocarcinoma (AGC). The results from clinical trials of various PD-L1 antibody clones are variable and the exchangeability of these assays is a highly sought goal. The aim of this study was to determine whether three different PD-L1 assays (SP263 and 22C3 on the Dako and Ventana platforms) are interchangeable through analysis of their concordance rate within samples between biopsy and paired resected specimens. One hundred pairs of biopsied and resected AGC specimens were collected and stained for PD-L1. The combined positive score (CPS) was used for the IHC analysis and a four tiered system was applied, i.e., <1, 1 to < 5, 5 to 50, and >50. The agreement for the different IHC assays was low across all cut-offs with the biopsied or resected specimens (biopsy, κ=0.17-0.453; resection, κ=0.02-0.311). The overall positive agreement (OPA) for the PD-L1 results from the biopsy and resection tissues was 100% (SP263, κ=1), 86% (22C3 on the Dako platform, κ=0.693) and 93% (22C3 on the Ventana platform, κ=0.82) at the CPS1 cut-off. The low concordances among the three PD-L1 IHC assays indicated that they cannot be used interchangeably in clinical practice. The results of the SP263 assay using CPS1 showed the highest agreement between the biopsy and resection specimens, suggesting SP263 may provide the most representative approach for the evaluation of PD-L1 status in gastric cancer.
Collapse
|
5
|
Chung HC, Bang YJ, S Fuchs C, Qin SK, Satoh T, Shitara K, Tabernero J, van Cutsem E, Alsina M, Cao ZA, Lu J, Bhagia P, Shih CS, Janjigian YY. First-line pembrolizumab/placebo plus trastuzumab and chemotherapy in HER2-positive advanced gastric cancer: KEYNOTE-811. Future Oncol 2021; 17:491-501. [PMID: 33167735 PMCID: PMC8411394 DOI: 10.2217/fon-2020-0737] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022] Open
Abstract
Treatment options for patients with HER2-positive advanced gastric cancer are limited, and the prognosis for these patients is poor. Pembrolizumab has demonstrated promising antitumor activity in patients with advanced gastric or gastroesophageal junction adenocarcinoma as monotherapy, in combination with chemotherapy and in combination with trastuzumab. Combining pembrolizumab with trastuzumab and chemotherapy may therefore provide a benefit for patients with advanced HER2-positive gastric cancer. Here we aimed to describe the design of and rationale for the randomized, double-blind, placebo-controlled Phase III KEYNOTE-811 study, which will evaluate the efficacy and safety of pembrolizumab or placebo in combination with trastuzumab and chemotherapy as first-line treatment for patients with advanced HER2-positive gastric or gastroesophageal junction adenocarcinoma. Clinical trial registration: NCT03615326 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Hyun Cheol Chung
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Yung-Jue Bang
- Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Charles S Fuchs
- Yale Cancer Center, Smilow Cancer Hospital, New Haven, CT 06510, USA
| | - Shu-Kui Qin
- Cancer Center of People's Liberation Army, Nanjing, 210002, China
| | - Taroh Satoh
- Osaka University, Suita, Osaka, 565-0871, Japan
| | - Kohei Shitara
- National Cancer Center Hospital East, Kashiwa, 277-8577, Japan
| | - Josep Tabernero
- Vall d'Hebron University Hospital & Institute of Oncology, Barcelona, 08035, Spain
| | - Eric van Cutsem
- University Hospitals Gasthuisberg Leuven & KU Leuven, Leuven, 03001, Belgium
| | - Maria Alsina
- Vall d'Hebron University Hospital & Institute of Oncology, Barcelona, 08035, Spain
| | - Zhu Alexander Cao
- Department of Medical Oncology, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Jia Lu
- Department of Medical Oncology, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Pooja Bhagia
- Department of Medical Oncology, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Chie-Schin Shih
- Department of Medical Oncology, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | | |
Collapse
|
6
|
Kim W, Chu TH, Nienhüser H, Jiang Z, Portillo AD, Remotti HE, White RA, Hayakawa Y, Tomita H, Fox JG, Drake CG, Wang TC. PD-1 Signaling Promotes Tumor-Infiltrating Myeloid-Derived Suppressor Cells and Gastric Tumorigenesis in Mice. Gastroenterology 2021; 160:781-796. [PMID: 33129844 PMCID: PMC7878361 DOI: 10.1053/j.gastro.2020.10.036] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Immune checkpoint inhibitors have limited efficacy in many tumors. We investigated mechanisms of tumor resistance to inhibitors of programmed cell death-1 (PDCD1, also called PD-1) in mice with gastric cancer, and the role of its ligand, PD-L1. METHODS Gastrin-deficient mice were given N-methyl-N-nitrosourea (MNU) in drinking water along with Helicobacter felis to induce gastric tumor formation; we also performed studies with H/K-ATPase-hIL1B mice, which develop spontaneous gastric tumors at the antral-corpus junction and have parietal cells that constitutively secrete interleukin 1B. Mice were given injections of an antibody against PD-1 or an isotype control before tumors developed, or anti-PD-1 and 5-fluorouracil and oxaliplatin, or an antibody against lymphocyte antigen 6 complex locus G (also called Gr-1), which depletes myeloid-derived suppressor cells [MDSCs]), after tumors developed. We generated knock-in mice that express PD-L1 specifically in the gastric epithelium or myeloid lineage. RESULTS When given to gastrin-deficient mice before tumors grew, anti-PD-1 significantly reduced tumor size and increased tumor infiltration by T cells. However, anti-PD-1 alone did not have significant effects on established tumors in these mice. Neither early nor late anti-PD-1 administration reduced tumor growth in the presence of MDSCs in H/K-ATPase-hIL-1β mice. The combination of 5-fluorouracil and oxaliplatin reduced MDSCs, increased numbers of intra-tumor CD8+ T cells, and increased the response of tumors to anti-PD-1; however, this resulted in increased tumor expression of PD-L1. Expression of PD-L1 by tumor or immune cells increased gastric tumorigenesis in mice given MNU. Mice with gastric epithelial cells that expressed PD-L1 did not develop spontaneous tumors, but they developed more and larger tumors after administration of MNU and H felis, with accumulation of MDSCs. CONCLUSIONS In mouse models of gastric cancer, 5-fluorouracil and oxaliplatin reduce numbers of MDSCs to increase the effects of anti-PD-1, which promotes tumor infiltration by CD8+ T cells. However, these chemotherapeutic agents also induce expression of PD-L1 by tumor cells. Expression of PD-L1 by gastric epithelial cells increases tumorigenesis in response to MNU and H felis, and accumulation of MDSCs, which promote tumor progression. The timing and site of PD-L1 expression is therefore important in gastric tumorigenesis and should be considered in design of therapeutic regimens.
Collapse
Affiliation(s)
- Woosook Kim
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Timothy H. Chu
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Henrik Nienhüser
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Zhengyu Jiang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Armando Del Portillo
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Helen E. Remotti
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Ruth A. White
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - James G. Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 01239, USA
| | - Charles G. Drake
- Division of Hematology and Oncology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Timothy C. Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
7
|
Abstract
BACKGROUND PD-1/PD-L1-Immunotherapy has been approved for gastric carcinoma. PD-L1 assessment by immunohistochemistry is the principle biomarker. Are biopsies able to map the actual PD-L1 status of the entire tumor? METHODS Whole tumor slides of 56 gastric carcinoma were analyzed to determine the distribution of PD-L1 positive cells in the entire tumor areas. Tissue micro arrays with four cores of the tumor surface, which represents the endoscopically accessible biopsy zone, were built from the same tumors. The PD-L1 CPS value was determined separately for each core. Preoperative diagnostic biopsies were available for 22 of the tumors. PD-L1 prevalence, sensitivity and specificity were analyzed using the whole tumor slides as reference scores. Molecular subtyping was performed and related to the PD-L1 status. RESULTS 27.3% of cases were PD-L1 negative (CPS < 1), 43.6% showed low PD-L1 expression (CPS ≥ 1 to < 5), 12.7% moderate (CPS ≥ 5 to < 10) and 16.4% strong expression (CPS ≥ 10). The biopsies showed best test characteristics if four surface biopsies were analyzed combined, i.e., the CPS was calculated across all four biopsies. The prevalence showed a distribution similar to the resection specimens, sensitivity was 0.73 and specificity 1.0. Using fewer surface biopsies decreased sensitivity and specificity and caused false-negative classifications. Compared to the TMAs, the preoperative biopsies showed reduced sensitivity (0.412). CONCLUSIONS This is the first comprehensive study to optimize PD-L1 assessment in gastric cancer using endoscopically available tissue. The obtained PD-L1 prevalence is consistent with data of current clinical studies. Calculation of the test characteristics shows that surface biopsies can be indicative of the true PD-L1 status based on the resection specimen. However, an adequate number of biopsies is required. In this study, n = 4 biopsies yielded best results.
Collapse
|