1
|
KURILO TATIANA, PENTZ REBECCAD. Comparative analysis of breast and lung cancer survival rates and clinical trial enrollments among rural and urban patients in Georgia. Oncol Res 2024; 32:1401-1406. [PMID: 39220122 PMCID: PMC11361898 DOI: 10.32604/or.2024.050266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/16/2024] [Indexed: 09/04/2024] Open
Abstract
Objectives Rural patients have poor cancer outcomes and clinical trial (CT) enrollment compared to urban patients due to attitudinal, awareness, and healthcare access differential. Knowledge of cancer survival disparities and CT enrollment is important for designing interventions and innovative approaches to address the stated barriers. The study explores the potential disparities in cancer survival rates and clinical trial enrollments in rural and urban breast and lung cancer patients. Our hypotheses are that for both cancer types, urban cancer patients will have longer 5-year survival rates and higher enrollment rates in clinical trials than those in rural counties. Methods We compared breast and lung cancer patients' survival rates and enrollment ratios in clinical trials between rural (RUCC 4-9) and urban counties in Georgia at a Comprehensive Cancer Center (CCC). To assess these differences, we carried out a series of independent samples t-tests and Chi-Square tests. Results The outcomes indicate comparable 5-year survival rates across rural and urban counties for breast and lung cancer patients, failing to substantiate our hypothesis. While clinical trial enrollment rates demonstrated a significant difference between breast and lung cancer patients at CCC, no significant variation was observed based on rural or urban classification. Conclusion These findings underscore the need for further research into the representation of rural patients with diverse cancer types at CCC and other cancer centers. Further, the findings have considerable implications for the initiation of positive social change to improve CT participation and reduce cancer survival disparities.
Collapse
Affiliation(s)
- TATIANA KURILO
- Winship Cancer Institute, Emory University School of Medicine, Emory University, Atlanta, 30322, USA
| | - REBECCA D. PENTZ
- Winship Cancer Institute, Emory University School of Medicine, Emory University, Atlanta, 30322, USA
| |
Collapse
|
2
|
Jia G, Wang Y, Wang J, Yu B, Zhao H, Zhao Z, Zhao W, Gao Y, Wang B, Song Z. Benzimidazole-based structure optimization to discover novel anti-gastric cancer agents targeting ROS/MAPK pathway. J Biochem Mol Toxicol 2024; 38:e23762. [PMID: 38967723 DOI: 10.1002/jbt.23762] [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: 01/31/2024] [Revised: 04/22/2024] [Accepted: 06/24/2024] [Indexed: 07/06/2024]
Abstract
Given the malignancy of gastric cancer, developing highly effective and low-toxic targeted drugs is essential to prolong patient survival and improve patient outcomes. In this study, we conducted structural optimizations based on the benzimidazole scaffold. Notably, compound 8 f presented the most potent antiproliferative activity in MGC803 cells and induced cell cycle arrest at the G0/G1 phase. Further mechanistic studies demonstrated that compound 8 f caused the apoptosis of MGC803 cells by elevating intracellular reactive oxygen species (ROS) levels and activating the mitogen-activated protein kinase (MAPK) signaling pathway, accompanied by corresponding markers change. In vivo investigations additionally validated the inhibitory effect of compound 8 f on tumor growth in xenograft models bearing MGC803 cells without obvious toxicity. Our studies suggest that compound 8 f holds promise as a potential and safe lead compound for developing anti-gastric cancer agents.
Collapse
Affiliation(s)
- Gang Jia
- Department of Oncology, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuanying Wang
- Academy of Medical Sciences; People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Jikuan Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Bingxin Yu
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haiyang Zhao
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ze Zhao
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenming Zhao
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ya Gao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Bo Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhiyu Song
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
3
|
Morgagni P, Bencivenga M, Carneiro F, Cascinu S, Derks S, Di Bartolomeo M, Donohoe C, Eveno C, Gisbertz S, Grimminger P, Gockel I, Grabsch H, Kassab P, Langer R, Lonardi S, Maltoni M, Markar S, Moehler M, Marrelli D, Mazzei MA, Melisi D, Milandri C, Moenig PS, Mostert B, Mura G, Polkowski W, Reynolds J, Saragoni L, Van Berge Henegouwen MI, Van Hillegersberg R, Vieth M, Verlato G, Torroni L, Wijnhoven B, Tiberio GAM, Yang HK, Roviello F, de Manzoni G. International consensus on the management of metastatic gastric cancer: step by step in the foggy landscape : Bertinoro Workshop, November 2022. Gastric Cancer 2024; 27:649-671. [PMID: 38634954 PMCID: PMC11193703 DOI: 10.1007/s10120-024-01479-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 02/05/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Many gastric cancer patients in Western countries are diagnosed as metastatic with a median overall survival of less than twelve months using standard chemotherapy. Innovative treatments, like targeted therapy or immunotherapy, have recently proved to ameliorate prognosis, but a general agreement on managing oligometastatic disease has yet to be achieved. An international multi-disciplinary workshop was held in Bertinoro, Italy, in November 2022 to verify whether achieving a consensus on at least some topics was possible. METHODS A two-round Delphi process was carried out, where participants were asked to answer 32 multiple-choice questions about CT, laparoscopic staging and biomarkers, systemic treatment for different localization, role and indication of palliative care. Consensus was established with at least a 67% agreement. RESULTS The assembly agreed to define oligometastases as a "dynamic" disease which either regresses or remains stable in response to systemic treatment. In addition, the definition of oligometastases was restricted to the following sites: para-aortic nodal stations, liver, lung, and peritoneum, excluding bones. In detail, the following conditions should be considered as oligometastases: involvement of para-aortic stations, in particular 16a2 or 16b1; up to three technically resectable liver metastases; three unilateral or two bilateral lung metastases; peritoneal carcinomatosis with PCI ≤ 6. No consensus was achieved on how to classify positive cytology, which was considered as oligometastatic by 55% of participants only if converted to negative after chemotherapy. CONCLUSION As assessed at the time of diagnosis, surgical treatment of oligometastases should aim at R0 curativity on the entire disease volume, including both the primary tumor and its metastases. Conversion surgery was defined as surgery on the residual volume of disease, which was initially not resectable for technical and/or oncological reasons but nevertheless responded to first-line treatment.
Collapse
Affiliation(s)
- Paolo Morgagni
- Department of General Surgery, Morgagni-Pierantoni Hospital, Forlì, Italy
| | - Maria Bencivenga
- General and Upper GI Surgery, Department of Surgery, University Hospital Verona, University of Verona, Verona, Italy.
| | - Fatima Carneiro
- Department of Pathology, Centro Hospitalar de São João, Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), Porto, Portugal
| | - Stefano Cascinu
- Department of Medical Oncology, Comprehensive Cancer Center, Università Vita-Salute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Sarah Derks
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Maria Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Claire Donohoe
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Clarisse Eveno
- Department of Digestive and Oncologic Surgery, Claude Huriez University Hospital, Centre Hospitalier Universitaire (CHU) Lille, Université de Lille, Lille, France
| | - Suzanne Gisbertz
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Peter Grimminger
- Department of General, Visceral and Transplant Surgery, University Medical Center, University of Mainz, Mainz, Germany
| | - Ines Gockel
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Heike Grabsch
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Paulo Kassab
- Gastric Surgery Division, BP Gastric Surgery Department, Santa Casa Medical School, São Paulo, Brazil
| | - Rupert Langer
- Institute of Pathology and Microbiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040, Linz, Austria
| | - Sara Lonardi
- Istituto Oncologico Veneto IOV-IRCCS, Padua, Italy
| | - Marco Maltoni
- Unit of Palliative Care, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Sheraz Markar
- Surgical Interventional Trials Unit, University of Oxford, Oxford, UK
| | - Markus Moehler
- Department of Medicine, Johannes-Gutenberg University Clinic, Mainz, Germany
| | - Daniele Marrelli
- Unit of General Surgery and Surgical Oncology, Department of Medicine Surgery and Neurosciences, University of Siena, 53100, Siena, Italy
| | - Maria Antonietta Mazzei
- Unit of Diagnostic Imaging, Department of Medical, Surgical and Neuro Sciences and of Radiological Sciences, Azienda Ospedaliero-Universitaria Senese, University of Siena, 53100, Siena, Italy
| | - Davide Melisi
- Medical Oncology at the Department of Medicine, University of Verona, Verona, Italy
| | - Carlo Milandri
- Department of Oncology, San Donato Hospital, 52100, Arezzo, Italy
| | | | - Bianca Mostert
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Gianni Mura
- Department of Surgery, San Donato Hospital, Arezzo, Italy
| | - Wojciech Polkowski
- Department of Surgical Oncology, Medical University of Lublin, Radziwiłłowska 13 St, 20-080, Lublin, Poland
| | | | - Luca Saragoni
- Pathology Unit, Santa Maria delle Croci Ravenna Hospital, Ravenna, Italy
| | - Mark I Van Berge Henegouwen
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Michael Vieth
- Institute of Pathology, Klinikum Bayreuth, Bayreuth, Germany
| | - Giuseppe Verlato
- Department of Diagnostics and Public Health, Section of Epidemiology and Medical Statistics, University of Verona, Verona, Italy
| | - Lorena Torroni
- Department of Diagnostics and Public Health, Section of Epidemiology and Medical Statistics, University of Verona, Verona, Italy
| | - Bas Wijnhoven
- Department of Surgery, Erasmus MC-University Medical Centre Rotterdam, Rotterdam, Netherlands
| | | | - Han-Kwang Yang
- Surgical Department, SNUH National Cancer Center, Seoul, Korea
| | - Franco Roviello
- Unit of General Surgery and Surgical Oncology, Department of Medicine Surgery and Neurosciences, University of Siena, 53100, Siena, Italy
| | - Giovanni de Manzoni
- General and Upper GI Surgery, Department of Surgery, University Hospital Verona, University of Verona, Verona, Italy
| |
Collapse
|
4
|
He J, Yi J, Ji L, Dai L, Chen Y, Xue W. ECHDC2 inhibits the proliferation of gastric cancer cells by binding with NEDD4 to degrade MCCC2 and reduce aerobic glycolysis. Mol Med 2024; 30:69. [PMID: 38783226 PMCID: PMC11118108 DOI: 10.1186/s10020-024-00832-9] [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: 01/04/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND The Enoyl-CoA hydratase/isomerase family plays a crucial role in the metabolism of tumors, being crucial for maintaining the energy balance and biosynthetic needs of cancer cells. However, the enzymes within this family that are pivotal in gastric cancer (GC) remain unclear. METHODS We employed bioinformatics techniques to identify key Enoyl-CoA hydratase/isomerase in GC. The expression of ECHDC2 and its clinical significance were validated through tissue microarray analysis. The role of ECHDC2 in GC was further assessed using colony formation assays, CCK8 assay, EDU assay, Glucose and lactic acid assay, and subcutaneous tumor experiments in nude mice. The mechanism of action of ECHDC2 was validated through Western blotting, Co-immunoprecipitation, and immunofluorescence experiments. RESULTS Our analysis of multiple datasets indicates that low expression of ECHDC2 in GC is significantly associated with poor prognosis. Overexpression of ECHDC2 notably inhibits aerobic glycolysis and proliferation of GC cells both in vivo and in vitro. Further experiments revealed that overexpression of ECHDC2 suppresses the P38 MAPK pathway by inhibiting the protein level of MCCC2, thereby restraining glycolysis and proliferation in GC cells. Ultimately, it was discovered that ECHDC2 promotes the ubiquitination and subsequent degradation of MCCC2 protein by binding with NEDD4. CONCLUSIONS These findings underscore the pivotal role of the ECHDC2 in regulating aerobic glycolysis and proliferation in GC cells, suggesting ECHDC2 as a potential therapeutic target in GC.
Collapse
Affiliation(s)
- Jiancheng He
- Department of Gastrointestinal Surgery, Affliated Hospital of Nantong University, Medical School of Nantong University, 20 Xisi Street, Nantong, 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Nantong Key Laboratory of Gastrointestinal Oncology, Nantong, 226001, China
| | - Jianfeng Yi
- Department of Gastrointestinal Surgery, Affliated Hospital of Nantong University, Medical School of Nantong University, 20 Xisi Street, Nantong, 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Nantong Key Laboratory of Gastrointestinal Oncology, Nantong, 226001, China
| | - Li Ji
- Department of Gastrointestinal Surgery, Affliated Hospital of Nantong University, Medical School of Nantong University, 20 Xisi Street, Nantong, 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Nantong Key Laboratory of Gastrointestinal Oncology, Nantong, 226001, China
| | - Lingchen Dai
- Department of Gastrointestinal Surgery, Affliated Hospital of Nantong University, Medical School of Nantong University, 20 Xisi Street, Nantong, 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Nantong Key Laboratory of Gastrointestinal Oncology, Nantong, 226001, China
| | - Yu Chen
- Department of Gastrointestinal Surgery, Affliated Hospital of Nantong University, Medical School of Nantong University, 20 Xisi Street, Nantong, 226001, China.
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China.
- Nantong Key Laboratory of Gastrointestinal Oncology, Nantong, 226001, China.
| | - Wanjiang Xue
- Department of Gastrointestinal Surgery, Affliated Hospital of Nantong University, Medical School of Nantong University, 20 Xisi Street, Nantong, 226001, China.
- Nantong Key Laboratory of Gastrointestinal Oncology, Nantong, 226001, China.
| |
Collapse
|
5
|
Lin G, Liu Z, Shang‐Guan Z, Zeng G, Lin J, Wu J, Chen Q, Xie J, Li P, Huang C, Zheng C. Comparison of the efficacy between immunochemotherapy and chemotherapy in gastric cancer accompanied with synchronous liver metastases: A real-world retrospective study. Cancer Med 2023; 12:12221-12233. [PMID: 37062073 PMCID: PMC10278523 DOI: 10.1002/cam4.5917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/20/2023] [Accepted: 03/27/2023] [Indexed: 04/17/2023] Open
Abstract
BACKGROUND Few studies have investigated the efficacy of comprehensive therapies, including immunotherapy, for gastric cancer with synchronous liver metastases (GCLM). We retrospectively compared the effect of immunochemotherapy and chemotherapy alone as conversion therapies on the oncological outcomes of patients with GCLM. METHODS The clinicopathological data of 100 patients with GCLM from February 2017 to October 2021 at our institution were retrospectively analyzed. Patients were divided into immunochemotherapy (n = 33) and chemotherapy-alone (n = 67) groups. RESULTS Baseline clinicopathological data did not differ significantly between the two groups. The immunochemotherapy group had a higher overall response rate (59.4% vs. 44.0%, p = 0.029) and disease control rate (71.9% vs. 49.2%, p = 0.036) than the chemotherapy group. The immunochemotherapy group showed better tumor regression in the gastric mass, metastatic lymph nodes, and liver lesions than the chemotherapy group. Ten (30.3%) patients in the immunochemotherapy group and 13 (19.4%) patients in the chemotherapy group underwent surgery after conversion therapy. However, the difference was not statistically significant. The overall survival (OS) and progression-free survival (PFS) rates were better in the immunochemotherapy group than in the chemotherapy group. Treatment-related adverse events occurred in 24 (72.7%) and 47 (70.1%) patients in the immunochemotherapy and chemotherapy groups, respectively. CONCLUSIONS As a conversion therapy for GCLM, immunotherapy yielded better primary and metastatic tumor regression and survival benefits, with no increase in adverse events compared to chemotherapy.
Collapse
Affiliation(s)
- Guang‐Tan Lin
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Zhi‐Yu Liu
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Zhi‐Xin Shang‐Guan
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Gui‐Rong Zeng
- Fujian Medical University Diagnostic Pathology CenterFujian Medical UniversityFuzhouChina
| | - Jian‐Xian Lin
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
- Key Laboratory of Ministry of Education of Gastrointestinal CancerFujian Medical UniversityFuzhouChina
| | - Ju Wu
- Department of General SurgeryAffiliated Zhongshan Hospital of Dalian UniversityDalianChina
| | - Qi‐Yue Chen
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Jian‐Wei Xie
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Ping Li
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
- Key Laboratory of Ministry of Education of Gastrointestinal CancerFujian Medical UniversityFuzhouChina
| | - Chang‐Ming Huang
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
- Key Laboratory of Ministry of Education of Gastrointestinal CancerFujian Medical UniversityFuzhouChina
| | - Chao‐Hui Zheng
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
- Key Laboratory of Ministry of Education of Gastrointestinal CancerFujian Medical UniversityFuzhouChina
| |
Collapse
|
6
|
Song Z, Zhao C, Yan J, Jiang D, Jia G. Carbenoxolone disodium suppresses the migration of gastric cancer by targeting HDAC6. Future Med Chem 2023; 15:333-344. [PMID: 36946221 DOI: 10.4155/fmc-2022-0246] [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] [Indexed: 03/23/2023] Open
Abstract
Aim: Because of the severe morbidity and mortality of gastric cancer, discovering new candidate drugs has been an urgent issue. The close association between histone deacetylase 6 (HDAC6) and gastric cancer makes the development of HDAC6-targeted anti-gastric cancer drugs a viable idea. Methods & results: Carbenoxolone disodium was identified as a novel HDAC6 inhibitor. Cellular thermal shift assay, surface plasmon resonance assay and molecular docking confirmed its binding ability to HDAC6. Cell viability, wound healing and transwell assays as well as animal studies have demonstrated that carbenoxolone disodium could block the proliferation and migration of gastric cancer cells MGC-803 in vitro and in vivo. Conclusion: This is the first report to indicate that carbenoxolone disodium could be an HDAC6 inhibitor with potential for treatment of gastric cancer.
Collapse
Affiliation(s)
- Zhiyu Song
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Chenglong Zhao
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Jingjing Yan
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Dandan Jiang
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Gang Jia
- Department of Oncology, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| |
Collapse
|
7
|
Chen S, Zhang H, Wei H, Tong Y, Chen X. Practical nomogram based on comprehensive CT texture analysis to preoperatively predict peritoneal occult metastasis of gastric cancer patients. Front Oncol 2022; 12:882584. [PMID: 36531010 PMCID: PMC9753569 DOI: 10.3389/fonc.2022.882584] [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: 02/24/2022] [Accepted: 10/24/2022] [Indexed: 04/16/2025] Open
Abstract
OBJECTIVES This study aims to evaluate whether a nomogram based on comprehensive CT texture analysis of primary tumor and peritoneotome combined with conventional CT signs can preoperatively predict peritoneal occult metastasis in gastric cancer patients. METHODS A total of 1,251 patients with gastric cancer (GC) were retrospectively analyzed in Fujian Province Hospital between 2008 and 2020. Patients from the occult peritoneal metastasis (PM) group were initially diagnosed as PM-negative on CT and later confirmed as PM-positive through laparoscopy or surgery. The group without PM was randomly sampled from patients without PM. The preoperative CT signs and texture features and clinical characteristics of patients were retrospectively analyzed. Hazard factors of occult PM were identified by univariate analysis and multivariate logistic regression analysis, which were intended for creating prediction models. A nomogram was established based on the model with the highest predictive efficacy and clinical application value. RESULTS A total of 31 patients with occult PM and 165 patients without PM were enrolled in this study. The maximum size, thickness, enhancement, serous involvement of primary GC tumor and ascites on CT, and texture features such as inhomogeneity of the primary tumor, standard deviation, and inhomogeneity of the peritoneum were determined as independent predictors that could be jointly applied to predict occult PM. We separately constructed five forecast models using CT signs, primary tumor texture, peritoneum texture, primary tumor texture + peritoneum texture, and their combination for predicting occult PM. These five prediction models achieved an AUC value of 0.832, 0.70, 0.784, 0.838, and 0.941, respectively. The DeLong test and Decision Curve Analysis (DCA) showed that the joint model, containing three meaningful CT signs (maximum size, thickness, and ascites) and two meaningful texture parameters (inhomogeneity of the primary tumor and inhomogeneity of the peritoneum), possessed the best predictive performance and clinical application (p<0.05). A forecast nomogram was subsequently established from the model above-mentioned. The calibration curves of the nomogram indicated a good consistency (a concordance index of 0.807) between the projection and the actual observation of occult PM. CONCLUSIONS A practical projection nomogram based on the comprehensive CT texture analysis of a primary tumor and peritoneotome combined with conventional CT signs was constructed in our study, which can be conveniently used in preoperative personalized prediction of occult PM for GC patients, and acts as a recommendation for the optimization of clinical management.
Collapse
Affiliation(s)
- Shuxiang Chen
- Department of Radiology, Provincial Clinical College of Fujian Medical University, Fujian Province Hospital, Fuzhou, China
| | - Huijuan Zhang
- Department of Radiology, Provincial Clinical College of Fujian Medical University, Fujian Province Hospital, Fuzhou, China
| | - Hong Wei
- Department of Cadre Health Care Office, Provincial Clinical College of Fujian Medical University, Fujian Province Hospital, Fuzhou, China
| | - Yongxiu Tong
- Department of Radiology, Provincial Clinical College of Fujian Medical University, Fujian Province Hospital, Fuzhou, China
| | - Xiaofang Chen
- Department of Radiology, Provincial Clinical College of Fujian Medical University, Fujian Province Hospital, Fuzhou, China
| |
Collapse
|
8
|
Nie C, Xu W, Lv H, Gao X, Li G, Chen B, Wang J, Liu Y, Zhao J, He Y, Wang S, Chen X. Tailoring second-line or above therapy for patients with advanced or metastatic gastric cancer: A multicenter real-world study. Front Pharmacol 2022; 13:1043217. [DOI: 10.3389/fphar.2022.1043217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/08/2022] [Indexed: 11/18/2022] Open
Abstract
Background: There is currently still a lack of effective therapeutic manner after the failure of first-line therapy for patients with advanced or metastatic gastric cancer. The present study aimed to evaluate the clinical efficacy and safety of different treatment strategies as second-line or above therapy for patients with advanced or metastatic gastric cancer.Methods: This was an observational multicenter real-world study. From January 2018 to December 2020, advanced or metastatic gastric cancer patients who have failed prior therapy were enrolled and treated with chemotherapy, anti-angiogenic TKIs (tyrosine kinase inhibitors) + chemotherapy or TKIs + ICIs (immune checkpoint inhibitors). In this study, progression free survival (PFS) was the primary end-point. Other evaluation indicators were objective response rate (ORR), disease control rate (DCR), overall survival (OS) and drug toxicities.Results: 162 patients were enrolled, of which 61 patients received chemotherapy, 47 patients received TKIs plus chemotherapy, and 54 patients received TKIs + ICIs. No statistically significant difference existed in ORR among groups (16.4% vs. 19.1% vs. 18.5%, p = 0.924). Patients who received TKIs plus chemotherapy obtained better DCR compared with the chemotherapy group (78.7% vs. 54.1%, p = 0.008), and simultaneously, the median PFS (3.3 m vs. 2.8 m, p = 0.001) and OS (8.0 m vs. 5.8 m, p = 0.005) in TKIs plus chemotherapy group were superior to chemotherapy group. Consistent results were observed in subgroup analysis, including sex, age, ECOG, number of metastatic sites and treatment line. No statistically differences were found between TKIs + ICIs and the chemotherapy group concerning DCR (63.0% vs. 54.1%, p = 0.336), median PFS (3.0 m vs. 2.8 m, p = 0.051) and OS (5.2 m vs. 5.8 m, p = 0.260). Different treatment manner present a special spectrum of adverse events (AEs), and the incidence of Grade 3–4 AEs were 31.1%, 38.3% and 18.5%, respectively.Conclusion: Compared with chemotherapy, anti-angiogenic TKIs plus chemotherapy demonstrated superior second-line or above therapeutic efficacy for advanced or metastatic gastric cancer with well tolerated toxicity. However, TKIs + ICIs failed to demonstrate a clinical advantage over chemotherapy.
Collapse
|
9
|
Ma N, Qiao H, Tao H, Gan X, Shan Z, Chen X, Zhou X. Treatment response, survival, and safety profile of camrelizumab plus apatinib regimen as third-line treatment in metastatic gastric cancer patients. Clin Res Hepatol Gastroenterol 2022; 46:101962. [PMID: 35636681 DOI: 10.1016/j.clinre.2022.101962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/22/2022] [Accepted: 05/25/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Camrelizumab, as a PD-1 inhibitor on the market recently, presents favorable therapeutic efficacy in several advanced cancers, while its application in metastatic gastric cancer (mGC) lacks data. This study aimed to assess treatment response, survival profile, and adverse events of camrelizumab plus apatinib regimen as third-line treatment in mGC patients. METHODS Nineteen mGC patients who received camrelizumab plus apatinib as third-line treatment were analyzed in this observational study. Subsequently, treatment response and adverse events were documented, then progression-free survival (PFS) and overall survival (OS) were calculated. RESULTS No (0.0%) patient achieved complete response; 5 (26.3%) patients achieved partial response; 8 (42.1%) patients had stable disease; 6 (31.6%) patients had progressive disease, resulting in objective response rate and disease control rate of 26.3% and 68.4%, respectively. Meanwhile, the median PFS and OS were 7.0 (95%CI: 2.9-11.0) months and 10.0 (95%CI: 7.4-12.6) months, accordingly. Besides, multiple metastases linked with worse PFS (P = 0.029) and OS (P = 0.021); Eastern Cooperative Oncology Group performance status (ECOG PS) score 1 (vs. 0) related to shorter OS (P = 0.030). Worth noting, the common adverse events were fatigue (42.1%), anemia (42.1%), neutropenia (42.1%), leukopenia (36.8%), pruritus (31.6%), proteinuria (31.6%), nausea and vomiting (31.6%), reactive capillary hemangioma (31.6%) and thrombocytopenia (31.6%). Meanwhile, grade 3-4 adverse events only included: thrombocytopenia (5.3%), hypertension (5.3%), and proteinuria (5.3%). CONCLUSION Camrelizumab plus apatinib as third-line treatment achieves satisfactory therapeutic efficacy and survival profile with generally manageable adverse events in mGC patients.
Collapse
Affiliation(s)
- Ning Ma
- Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 296 Shizi Street, Suzhou 215006, China; Department of General Surgery, Daqing Oilfield General Hospital, Daqing 163001, China
| | - Hui Qiao
- Department of Gastroenterology, Daqing Oilfield General Hospital, Daqing 163001, China
| | - Hanchuan Tao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 296 Shizi Street, Suzhou 215006, China
| | - Xinli Gan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 296 Shizi Street, Suzhou 215006, China
| | - Zhili Shan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 296 Shizi Street, Suzhou 215006, China
| | - Xiaomin Chen
- Department of General Surgery, Daqing Oilfield General Hospital, Daqing 163001, China
| | - Xiaojun Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 296 Shizi Street, Suzhou 215006, China.
| |
Collapse
|
10
|
Yu X, He S, Shen J, Huang Q, Yang P, Huang L, Pu D, Wang L, Li L, Liu J, Liu Z, Zhu L. Tumor vessel normalization and immunotherapy in gastric cancer. Ther Adv Med Oncol 2022; 14:17588359221110176. [PMID: 35872968 PMCID: PMC9297465 DOI: 10.1177/17588359221110176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 06/09/2022] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is a common malignant tumor, and patients with GC have a low survival rate due to limited effective treatment methods. Angiogenesis and immune evasion are two key processes in GC progression, and they act synergistically to promote tumor progression. Tumor vascular normalization has been shown to improve the efficacy of cancer immunotherapy, which in turn may be improved through enhanced immune stimulation. Therefore, it may be interesting to identify synergies between immunomodulatory agents and anti-angiogenic therapies in GC. This strategy aims to normalize the tumor microenvironment through the action of the anti-vascular endothelial growth factor while stimulating the immune response through immunotherapy and prolonging the survival of GC patients.
Collapse
Affiliation(s)
- Xianzhe Yu
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Shan He
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Jian Shen
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Qiushi Huang
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Peng Yang
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Lin Huang
- West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Dan Pu
- West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Li Wang
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Lu Li
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Jinghua Liu
- Department of Hepatobiliary Surgery, Linyi People's Hospital, Linyi, Shandong 276000, People's Republic of China
| | - Zelong Liu
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lingling Zhu
- Lung Cancer Center, West China Hospital of Sichuan University, No. 37, Guo Xue Xiang, Wuhou District, Chengdu, Sichuan 610041, People's Republic of China
| |
Collapse
|
11
|
Zhang J, Gao M, Niu Y, Sun J. From DNMT1 degrader to ferroptosis promoter: Drug repositioning of 6-Thioguanine as a ferroptosis inducer in gastric cancer. Biochem Biophys Res Commun 2022; 603:75-81. [DOI: 10.1016/j.bbrc.2022.03.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/05/2022] [Indexed: 02/09/2023]
|
12
|
Hu J, Yang S, Wang J, Zhang Q, Zhao L, Zhang D, Yu D, Jin M, Ma H, Liu H, Xue J, Zhang T. Blood alkaline phosphatase predicts prognosis of patients with advanced HER2-negative gastric cancer receiving immunotherapy. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1316. [PMID: 34532453 PMCID: PMC8422101 DOI: 10.21037/atm-21-3376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/11/2021] [Indexed: 12/02/2022]
Abstract
BACKGROUND Immune checkpoint blockade is effective against many cancer types, but few patients achieve a complete response (OR). Therefore, effective prognostic biomarkers are needed for metastatic gastric cancer (GC) patients after immune treatment. The present study assessed the value of hematological parameters as markers of the effectiveness of immune checkpoint blockade among metastatic GC patients. METHODS This retrospective study included patients with metastatic GC who underwent multiline chemotherapy including at least two courses of immunotherapy between September 2018 and December 2020. Patient and tumor characteristics were tested for prognostic significance by analysis of variance or chi-square test. Kaplan-Meier and Cox analyses were performed to identify factors associated with progression-free survival (PFS). RESULTS Sixty-one GC patients (mean age 55.61±11.97 years, range 23-80 years, 24 females, and 37 males) were included, and 27, 9 and 25 cases had organ only, peritoneum only, and simultaneous organ and peritoneum metastasis, respectively. Gastrectomy was performed in 24 cases, and there was no operative treatment in the other 37 cases, while all patients received two or more lines of chemotherapy. After immune treatment, 13 patients achieved a partial response (PR), 16 stable disease (SD), and 32 progressive disease (PD). The median PFS was 4.93±3.47 months. An alkaline phosphatase (ALP) level >225 U/L, a lactate dehydrogenase level (LDH) >299 U/L, and a body mass index (BMI) >24 kg/m2 were associated with a short PFS (P=0.01, P=0.008, and P=0.039, respectively). A Cox multivariate proportional hazard model indicated that higher ALP level was a significant prognostic indicator for adverse PFS. CONCLUSIONS Our data show an ALP cutoff of 225 U/L offered good prognostic sensitivity for HER2-negative metastatic GC. ALP measurement represents a convenient, cost-effective, and relatively sensitive screening tool, and prospective studies involving its evaluation in addition to other biomarkers in metastatic GC patients are indicated.
Collapse
Affiliation(s)
- Jianli Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengli Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuyue Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dejun Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dandan Yu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongli Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Xue
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Identification of pivotal genes associated with the prognosis of gastric carcinoma through integrated analysis. Biosci Rep 2021; 41:228128. [PMID: 33754626 PMCID: PMC8047542 DOI: 10.1042/bsr20203676] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Detecting and diagnosing gastric cancer (GC) during its early period remains greatly difficult. Our analysis was performed to detect core genes correlated with GC and explore their prognostic values. METHODS Microarray datasets from the Gene Expression Omnibus (GEO) (GSE54129) and The Cancer Genome Atlas (TCGA)-stomach adenocarcinoma (STAD) datasets were applied for common differentially co-expressed genes using differential gene expression analysis and Weighted Gene Co-expression Network Analysis (WGCNA). Functional enrichment analysis and protein-protein interaction (PPI) network analysis of differentially co-expressed genes were performed. We identified hub genes via the CytoHubba plugin. Prognostic values of hub genes were explored. Afterward, Gene Set Enrichment Analysis (GSEA) was used to analyze survival-related hub genes. Finally, the tumor-infiltrating immune cell (TIC) abundance profiles were estimated. RESULTS Sixty common differentially co-expressed genes were found. Functional enrichment analysis implied that cell-cell junction organization and cell adhesion molecules were primarily enriched. Hub genes were identified using the degree, edge percolated component (EPC), maximal clique centrality (MCC), and maximum neighborhood component (MNC) algorithms, and serpin family E member 1 (SERPINE1) was highly associated with the prognosis of GC patients. Moreover, GSEA demonstrated that extracellular matrix (ECM) receptor interactions and pathways in cancers were correlated with SERPINE1 expression. CIBERSORT analysis of the proportion of TICs suggested that CD8+ T cell and T-cell regulation were negatively associated with SERPINE1 expression, showing that SERPINE1 may inhibit the immune-dominant status of the tumor microenvironment (TME) in GC. CONCLUSIONS Our analysis shows that SERPINE1 is closely correlated with the tumorigenesis and progression of GC. Furthermore, SERPINE1 acts as a candidate therapeutic target and prognostic biomarker of GC.
Collapse
|
14
|
Ghidini M, Petrillo A, Botticelli A, Trapani D, Parisi A, La Salvia A, Sajjadi E, Piciotti R, Fusco N, Khakoo S. How to Best Exploit Immunotherapeutics in Advanced Gastric Cancer: Between Biomarkers and Novel Cell-Based Approaches. J Clin Med 2021; 10:1412. [PMID: 33915839 PMCID: PMC8037391 DOI: 10.3390/jcm10071412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Despite extensive research efforts, advanced gastric cancer still has a dismal prognosis with conventional treatment options. Immune checkpoint inhibitors have revolutionized the treatment landscape for many solid tumors. Amongst gastric cancer subtypes, tumors with microsatellite instability and Epstein Barr Virus positive tumors provide the strongest rationale for responding to immunotherapy. Various predictive biomarkers such as mismatch repair status, programmed death ligand 1 expression, tumor mutational burden, assessment of tumor infiltrating lymphocytes and circulating biomarkers have been evaluated. However, results have been inconsistent due to different methodologies and thresholds used. Clinical implementation therefore remains a challenge. The role of immune checkpoint inhibitors in gastric cancer is emerging with data from monotherapy in the heavily pre-treated population already available and studies in earlier disease settings with different combinatorial approaches in progress. Immune checkpoint inhibitor combinations with chemotherapy (CT), anti-angiogenics, tyrosine kinase inhibitors, anti-Her2 directed therapy, poly (ADP-ribose) polymerase inhibitors or dual checkpoint inhibitor strategies are being explored. Moreover, novel strategies including vaccines and CAR T cell therapy are also being trialed. Here we provide an update on predictive biomarkers for response to immunotherapy with an overview of their strengths and limitations. We discuss clinical trials that have been reported and trials in progress whilst providing an account of future steps needed to improve outcome in this lethal disease.
Collapse
Affiliation(s)
- Michele Ghidini
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | | | - Andrea Botticelli
- Department of Clinical and Molecular Medicine, Sapienza University, 00189 Rome, Italy;
- Medical Oncology (B), Policlinico Umberto I, 00161 Rome, Italy
| | - Dario Trapani
- Division of Early Drug Development for innovative therapies, European Institute of Oncology, IRCCS, 20141 Milan, Italy;
| | - Alessandro Parisi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
- Medical Oncology Unit, St. Salvatore Hospital, 67100 L’Aquila, Italy
| | - Anna La Salvia
- Department of Oncology, University Hospital 12 De Octubre, 28041 Madrid, Spain;
| | - Elham Sajjadi
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (E.S.); (R.P.); (N.F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Roberto Piciotti
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (E.S.); (R.P.); (N.F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Nicola Fusco
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (E.S.); (R.P.); (N.F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Shelize Khakoo
- Department of Medicine, Royal Marsden Hospital, London and Surrey, Sutton SM25PT, UK;
| |
Collapse
|
15
|
Zhang XH, Guo Q, Wang HY, Li YH, Khamis MY, Ma LY, Wang B, Liu HM. Gramine-based structure optimization to enhance anti-gastric cancer activity. Bioorg Chem 2021; 107:104549. [PMID: 33383324 DOI: 10.1016/j.bioorg.2020.104549] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 12/08/2020] [Indexed: 01/04/2023]
Abstract
Gramine is a natural indole alkaloid with a wide range of biological activities, but its anti-gastric cancer activity is poor. Herein, a pharmacophore fusion strategy was adopted to design and synthesize a new series of indole-azole hybrids on the structural basis of gramine. Based on our previous studies, different nitrogen-containing five-membered heterocyclic rings and terminal alkyne group were introduced into the indole-based scaffold to investigate their effect on improving the anti-gastric cancer activity of gramine derivatives. Structure-activity relationship (SAR) studies highlighted the role played by terminal alkyne in enhancing the inhibitory effect, and compound 16h displayed the best antiproliferative activity against gastric cancer MGC803 cells with IC50 value of 3.74 μM. Further investigations displayed compound 16h could induce mitochondria-mediated apoptosis, and caused cell cycle arrest at G2/M phase. Besides, compound 16h could inhibit the metastasis ability of MGC803 cells. Our studies may provide a new strategy for structural optimization of gramine to enhance anti-gastric cancer activity, and provide a potential candidate for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Xin-Hui Zhang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Qian Guo
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Heng-Ying Wang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yi-Han Li
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Mussa Yussuf Khamis
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Li-Ying Ma
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China; China Meheco Topfond Pharmaceutical Co., Ltd, PR China
| | - Bo Wang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Hong-Min Liu
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China.
| |
Collapse
|