1
|
Dinis-Ribeiro M, Libânio D, Uchima H, Spaander MCW, Bornschein J, Matysiak-Budnik T, Tziatzios G, Santos-Antunes J, Areia M, Chapelle N, Esposito G, Fernandez-Esparrach G, Kunovsky L, Garrido M, Tacheci I, Link A, Marcos P, Marcos-Pinto R, Moreira L, Pereira AC, Pimentel-Nunes P, Romanczyk M, Fontes F, Hassan C, Bisschops R, Feakins R, Schulz C, Triantafyllou K, Carneiro F, Kuipers EJ. Management of epithelial precancerous conditions and early neoplasia of the stomach (MAPS III): European Society of Gastrointestinal Endoscopy (ESGE), European Helicobacter and Microbiota Study Group (EHMSG) and European Society of Pathology (ESP) Guideline update 2025. Endoscopy 2025. [PMID: 40112834 DOI: 10.1055/a-2529-5025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
At a population level, the European Society of Gastrointestinal Endoscopy (ESGE), the European Helicobacter and Microbiota Study Group (EHMSG), and the European Society of Pathology (ESP) suggest endoscopic screening for gastric cancer (and precancerous conditions) in high-risk regions (age-standardized rate [ASR] > 20 per 100 000 person-years) every 2 to 3 years or, if cost-effectiveness has been proven, in intermediate risk regions (ASR 10-20 per 100 000 person-years) every 5 years, but not in low-risk regions (ASR < 10).ESGE/EHMSG/ESP recommend that irrespective of country of origin, individual gastric risk assessment and stratification of precancerous conditions is recommended for first-time gastroscopy. ESGE/EHMSG/ESP suggest that gastric cancer screening or surveillance in asymptomatic individuals over 80 should be discontinued or not started, and that patients' comorbidities should be considered when treatment of superficial lesions is planned.ESGE/EHMSG/ESP recommend that a high quality endoscopy including the use of virtual chromoendoscopy (VCE), after proper training, is performed for screening, diagnosis, and staging of precancerous conditions (atrophy and intestinal metaplasia) and lesions (dysplasia or cancer), as well as after endoscopic therapy. VCE should be used to guide the sampling site for biopsies in the case of suspected neoplastic lesions as well as to guide biopsies for diagnosis and staging of gastric precancerous conditions, with random biopsies to be taken in the absence of endoscopically suspected changes. When there is a suspected early gastric neoplastic lesion, it should be properly described (location, size, Paris classification, vascular and mucosal pattern), photodocumented, and two targeted biopsies taken.ESGE/EHMSG/ESP do not recommend routine performance of endoscopic ultrasonography (EUS), computed tomography (CT), magnetic resonance imaging (MRI), or positron emission tomography (PET)-CT prior to endoscopic resection unless there are signs of deep submucosal invasion or if the lesion is not considered suitable for endoscopic resection.ESGE/EHMSG/ESP recommend endoscopic submucosal dissection (ESD) for differentiated gastric lesions clinically staged as dysplastic (low grade and high grade) or as intramucosal carcinoma (of any size if not ulcerated or ≤ 30 mm if ulcerated), with EMR being an alternative for Paris 0-IIa lesions of size ≤ 10 mm with low likelihood of malignancy.ESGE/EHMSG/ESP suggest that a decision about ESD can be considered for malignant lesions clinically staged as having minimal submucosal invasion if differentiated and ≤ 30 mm; or for malignant lesions clinically staged as intramucosal, undifferentiated and ≤ 20 mm; and in both cases with no ulcerative findings.ESGE/EHMSG/ESP recommends patient management based on the following histological risk after endoscopic resection: Curative/very low-risk resection (lymph node metastasis [LNM] risk < 0.5 %-1 %): en bloc R0 resection; dysplastic/pT1a, differentiated lesion, no lymphovascular invasion, independent of size if no ulceration and ≤ 30 mm if ulcerated. No further staging procedure or treatment is recommended.Curative/low-risk resection (LNM risk < 3 %): en bloc R0 resection; lesion with no lymphovascular invasion and: a) pT1b, invasion ≤ 500 µm, differentiated, size ≤ 30 mm; or b) pT1a, undifferentiated, size ≤ 20 mm and no ulceration. Staging should be completed, and further treatment is generally not necessary, but a multidisciplinary discussion is required. Local-risk resection (very low risk of LNM but increased risk of local persistence/recurrence): Piecemeal resection or tumor-positive horizontal margin of a lesion otherwise meeting curative/very low-risk criteria (or meeting low-risk criteria provided that there is no submucosal invasive tumor at the resection margin in the case of piecemeal resection or tumor-positive horizontal margin for pT1b lesions [invasion ≤ 500 µm; well-differentiated; size ≤ 30 mm, and VM0]). Endoscopic surveillance/re-treatment is recommended rather than other additional treatment. High-risk resection (noncurative): Any lesion with any of the following: (a) a positive vertical margin (if carcinoma) or lymphovascular invasion or deep submucosal invasion (> 500 µm from the muscularis mucosae); (b) poorly differentiated lesions if ulceration or size > 20 mm; (c) pT1b differentiated lesions with submucosal invasion ≤ 500 µm with size > 30 mm; or (d) intramucosal ulcerative lesion with size > 30 mm. Complete staging and strong consideration for additional treatments (surgery) in multidisciplinary discussion.ESGE/EHMSG/ESP suggest the use of validated endoscopic classifications of atrophy (e. g. Kimura-Takemoto) or intestinal metaplasia (e. g. endoscopic grading of gastric intestinal metaplasia [EGGIM]) to endoscopically stage precancerous conditions and stratify the risk for gastric cancer.ESGE/EHMSG/ESP recommend that biopsies should be taken from at least two topographic sites (2 biopsies from the antrum/incisura and 2 from the corpus, guided by VCE) in two separate, clearly labeled vials. Additional biopsy from the incisura is optional.ESGE/EHMSG/ESP recommend that patients with extensive endoscopic changes (Kimura C3 + or EGGIM 5 +) or advanced histological stages of atrophic gastritis (severe atrophic changes or intestinal metaplasia, or changes in both antrum and corpus, operative link on gastritis assessment/operative link on gastric intestinal metaplasia [OLGA/OLGIM] III/IV) should be followed up with high quality endoscopy every 3 years, irrespective of the individual's country of origin.ESGE/EHMSG/ESP recommend that no surveillance is proposed for patients with mild to moderate atrophy or intestinal metaplasia restricted to the antrum, in the absence of endoscopic signs of extensive lesions or other risk factors (family history, incomplete intestinal metaplasia, persistent H. pylori infection). This group constitutes most individuals found in clinical practice.ESGE/EHMSG/ESP recommend H. pylori eradication for patients with precancerous conditions and after endoscopic or surgical therapy.ESGE/EHMSG/ESP recommend that patients should be advised to stop smoking and low-dose daily aspirin use may be considered for the prevention of gastric cancer in selected individuals with high risk for cardiovascular events.
Collapse
Affiliation(s)
- Mário Dinis-Ribeiro
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Gastroenterology Department, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Diogo Libânio
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Gastroenterology Department, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Hugo Uchima
- Endoscopy Unit Gastroenterology Department Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Endoscopy Unit, Teknon Medical Center, Barcelona, Spain
| | - Manon C W Spaander
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jan Bornschein
- Medical Research Council Translational Immune Discovery Unit (MRC TIDU), Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Tamara Matysiak-Budnik
- Department of Hepato-Gastroenterology & Digestive Oncology, Institut des Maladies de l'Appareil Digestif, Centre Hospitalier Universitaire de Nantes Nantes, France
- INSERM, Center for Research in Transplantation and Translational Immunology, University of Nantes, Nantes, France
| | - Georgios Tziatzios
- Agia Olga General Hospital of Nea Ionia Konstantopouleio, Athens, Greece
| | - João Santos-Antunes
- Gastroenterology Department, Centro Hospitalar S. João, Porto, Portugal
- Faculty of Medicine, University of Porto, Portugal
- University of Porto, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Instituto de Investigação e Inovação na Saúde (I3S), Porto, Portugal
| | - Miguel Areia
- Gastroenterology Department, Portuguese Oncology Institute of Coimbra (IPO Coimbra), Coimbra, Portugal
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), RISE@CI-IPO, (Health Research Network), Portuguese Institute of Oncology of Porto (IPO Porto), Porto, Portugal
| | - Nicolas Chapelle
- Department of Hepato-Gastroenterology & Digestive Oncology, Institut des Maladies de l'Appareil Digestif, Centre Hospitalier Universitaire de Nantes Nantes, France
- INSERM, Center for Research in Transplantation and Translational Immunology, University of Nantes, Nantes, France
| | - Gianluca Esposito
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Italy
| | - Gloria Fernandez-Esparrach
- Gastroenterology Department, ICMDM, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Spain
| | - Lumir Kunovsky
- 2nd Department of Internal Medicine - Gastroenterology and Geriatrics, University Hospital Olomouc, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
- Department of Surgery, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Gastroenterology and Digestive Endoscopy, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Mónica Garrido
- Gastroenterology Department, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Ilja Tacheci
- Gastroenterology, Second Department of Internal Medicine, University Hospital Hradec Kralove, Faculty of Medicine in Hradec Kralove, Charles University of Prague, Czech Republic
| | | | - Pedro Marcos
- Department of Gastroenterology, Pêro da Covilhã Hospital, Covilhã, Portugal
- Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Ricardo Marcos-Pinto
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), RISE@CI-IPO, (Health Research Network), Portuguese Institute of Oncology of Porto (IPO Porto), Porto, Portugal
- Gastroenterology Department, Centro Hospitalar do Porto, Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Leticia Moreira
- Gastroenterology Department, ICMDM, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Spain
| | - Ana Carina Pereira
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Pedro Pimentel-Nunes
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), RISE@CI-IPO, (Health Research Network), Portuguese Institute of Oncology of Porto (IPO Porto), Porto, Portugal
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto (FMUP), Portugal
- Gastroenterology and Clinical Research, Unilabs Portugal
| | - Marcin Romanczyk
- Department of Gastroenterology, Faculty of Medicine, Academy of Silesia, Katowice, Poland
- Endoterapia, H-T. Centrum Medyczne, Tychy, Poland
| | - Filipa Fontes
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Public Health and Forensic Sciences, and Medical Education Department, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Cesare Hassan
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Raf Bisschops
- Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
- Department of Translational Research in Gastrointestinal Diseases (TARGID), KU Leuven, Leuven, Belgium
| | - Roger Feakins
- Department of Cellular Pathology, Royal Free London NHS Foundation Trust, London, United Kingdom
- University College London, London, United Kingdom
| | - Christian Schulz
- Department of Medicine II, University Hospital, LMU Munich, Germany
| | - Konstantinos Triantafyllou
- Hepatogastroenterology Unit, Second Department of Internal Medicine-Propaedeutic, Medical School, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens, Greece
| | - Fatima Carneiro
- Institute of Molecular Pathology and Immunology at the University of Porto (IPATIMUP), Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- Pathology Department, Centro Hospitalar de São João and Faculty of Medicine, Porto, Portugal
| | - Ernst J Kuipers
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
2
|
Kristoff TJ, Evans S, Nayi P, Abousaud M, Goyal S, Liu Y, Shin D, Steuer CE, Saba NF, Schmitt NC. Statin Drugs Are Associated With Response to Immune Checkpoint Blockade in Recurrent/Metastatic Head and Neck Cancer. Cancer Med 2025; 14:e70718. [PMID: 40052634 PMCID: PMC11886884 DOI: 10.1002/cam4.70718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Statin drugs, frequently used to treat hyperlipidemia, are associated with improved survival outcomes in multiple solid tumor types, including head and neck squamous cell carcinoma (HNSCC). Preclinical studies suggest that manipulation of cholesterol with statins and other agents can enhance the function of multiple components involved in anti-tumor immune responses. Retrospective studies in other solid tumor types suggest that statin therapy is associated with improved responses to immune checkpoint blockade (ICB), but this has not yet been investigated in HNSCC. METHODS Pharmacy records were searched for patients with recurrent/metastatic HNSCC treated at our institution with pembrolizumab or nivolumab from 2015 to 2022. Patients who received less than 3 doses of ICB were excluded. Univariate and multivariate analyses were performed to determine the association between statin use and objective response, progression-free survival (PFS) and overall survival (OS). RESULTS A total of 158 patients were included. Statins were significantly associated with objective response; the strongest associations were seen with rosuvastatin and lovastatin. On multivariate analyses, statins were independently associated with objective response but not with PFS or OS. CONCLUSIONS Statin therapy appears to be an independent predictor of response to ICB in HNSCC. Larger, prospective studies are needed to determine whether specific statin drugs can improve survival outcomes in ICB-treated patients.
Collapse
Affiliation(s)
- Tyler J. Kristoff
- Department of Hematology and Medical OncologyEmory UniversityAtlantaGeorgiaUSA
- Winship Cancer Institute, Emory UniversityAtlantaGeorgiaUSA
| | - Sean Evans
- Department of Hematology and Medical OncologyEmory UniversityAtlantaGeorgiaUSA
- Winship Cancer Institute, Emory UniversityAtlantaGeorgiaUSA
| | - Pranay Nayi
- Children's Healthcare of AtlantaAtlantaGeorgiaUSA
| | - Marin Abousaud
- Astellas Pharma Global Development IncNorthbrookIllinoisUSA
| | - Subir Goyal
- Department of Biostatistics and BioinformaticsRollins School of Public Health of Emory UniversityAtlantaGeorgiaUSA
| | - Yuan Liu
- Department of Biostatistics and BioinformaticsRollins School of Public Health of Emory UniversityAtlantaGeorgiaUSA
| | - Dong Shin
- Department of Hematology and Medical OncologyEmory UniversityAtlantaGeorgiaUSA
- Winship Cancer Institute, Emory UniversityAtlantaGeorgiaUSA
| | - Conor E. Steuer
- Department of Hematology and Medical OncologyEmory UniversityAtlantaGeorgiaUSA
- Winship Cancer Institute, Emory UniversityAtlantaGeorgiaUSA
| | - Nabil F. Saba
- Department of Hematology and Medical OncologyEmory UniversityAtlantaGeorgiaUSA
- Winship Cancer Institute, Emory UniversityAtlantaGeorgiaUSA
| | - Nicole C. Schmitt
- Winship Cancer Institute, Emory UniversityAtlantaGeorgiaUSA
- Department of Otolaryngology – Head and Neck SurgeryEmory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
3
|
Holmberg D, Kauppila JH, Asplund J, Leijonmarck W, Mattsson F, Lagergren J. Statin use in relation to long-term survival after gastrectomy for gastric adenocarcinoma: a Swedish population-based cohort study. Gastric Cancer 2024; 27:590-597. [PMID: 38430275 PMCID: PMC11016510 DOI: 10.1007/s10120-024-01487-5] [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: 09/06/2023] [Accepted: 02/26/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Studies have suggested that medication with statins improves survival in patients with gastric cancer, but methodological issues have limited the interpretability and prohibited conclusive results. We aimed to provide valid evidence as to whether statin use improves survival of gastric adenocarcinoma. METHODS This nationwide and population-based cohort study included virtually all patients who underwent curatively intended surgery (gastrectomy) for gastric adenocarcinoma in Sweden between 2006 and 2015 with follow-up throughout 2019 for disease-specific mortality and 2020 for all-cause mortality. Data came from medical records and national healthcare registries. The exposure was statin use during the year prior to gastrectomy which was compared to no such use during the same period. The outcomes were 5-year disease-specific mortality (main) and 5-year all-cause mortality (secondary). Multivariable Cox regression provided hazard ratios (HR) with 95% confidence intervals (CI), adjusted for age, sex, education, calendar year, comorbidity, low-dose aspirin use, tumour sublocation, pathological tumour stage, neoadjuvant chemotherapy, annual surgeon volume, and surgical radicality. RESULTS Among 1515 participating patients, the mean age was 69 years and 58.4% were men. Statin use, identified in 399 (26.3%) patients, was not associated with any statistically significantly decreased 5-year disease-specific mortality (HR 0.99, 95% CI 0.82-1.21) or 5-year all-cause mortality (HR 0.94, 95% CI 0.79-1.12). No risk reductions were found across subgroups of age, sex, aspirin user status, or tumour stage, or in patients with long-term preoperative of postoperative use of statins, all with point estimates close to 1. CONCLUSIONS Perioperative use of statins does not seem to improve the 5-year survival in patients who undergo gastrectomy with curative intent for gastric adenocarcinoma in Sweden.
Collapse
Affiliation(s)
- Dag Holmberg
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital, Retzius Street 13A, 4th Floor, 171 77, Stockholm, Sweden.
| | - Joonas H Kauppila
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital, Retzius Street 13A, 4th Floor, 171 77, Stockholm, Sweden
- Department of Surgery, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Johannes Asplund
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital, Retzius Street 13A, 4th Floor, 171 77, Stockholm, Sweden
| | - Wilhelm Leijonmarck
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital, Retzius Street 13A, 4th Floor, 171 77, Stockholm, Sweden
| | - Fredrik Mattsson
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital, Retzius Street 13A, 4th Floor, 171 77, Stockholm, Sweden
| | - Jesper Lagergren
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital, Retzius Street 13A, 4th Floor, 171 77, Stockholm, Sweden
- School of Cancer and Pharmacological Sciences, King's College London, London, UK
| |
Collapse
|
4
|
Yang Y, Qi J, Hu J, Zhou Y, Zheng J, Deng W, Inam M, Guo J, Xie Y, Li Y, Xu C, Deng W, Chen W. Lovastatin/SN38 co-loaded liposomes amplified ICB therapeutic effect via remodeling the immunologically-cold colon tumor and synergized stimulation of cGAS-STING pathway. Cancer Lett 2024; 588:216765. [PMID: 38408604 DOI: 10.1016/j.canlet.2024.216765] [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: 11/28/2023] [Revised: 02/17/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
Current immune checkpoint blockade (ICB) immunotherapeutics have revolutionized cancer treatment. However, many cancers especially the "immunologically cold" tumors, do not respond to ICB, prompting the search for additional strategies to achieve durable responses. The cGAS-STING pathway, as an essential immune response pathway, has been demonstrated for a potent target to sensitize ICB immunotherapy. However, the low efficiency of conventional STING agonists limits their clinical application. Recent studies have shown that DNA topoisomerase I (TOPI) inhibitor chemodrug SN38 can activate the cGAS-STING pathway and induce an immune response through DNA damage, while the traditional statins medication lovastatin was found to inhibit DNA damage repair, which may in turn upregulate the damaged DNA level. Herein, we have developed a liposomal carrier co-loaded with SN38 and lovastatin (SL@Lip), which can be accumulated in tumors and efficiently released SN38 and lovastatin, addressing the problem of weak solubility of these two drugs. Importantly, lovastatin can increase DNA damage and enhance the activation of cGAS-STING pathway, coordinating with SN38 chemotherapy and exhibiting the enhanced combinational immunotherapy of PD-1 antibody by remodeling the tumor microenvironment in mouse colorectal cancer of both subcutaneous and orthotopic xenograft models. Overall, this study demonstrates that lovastatin-assisted cGAS-STING stimulation mediated by liposomal delivery system significantly strengthened both chemotherapy and immunotherapy of colorectal cancer, providing a clinically translational strategy for combinational ICB therapy in the "immunologically cold" tumors.
Collapse
Affiliation(s)
- Yi Yang
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Jialong Qi
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People's Hospital of Yunnan Province, Kunming, 650032, PR China
| | - Jialin Hu
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - You Zhou
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Jiena Zheng
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Wenxia Deng
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Muhammad Inam
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Jiaxin Guo
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yongyi Xie
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yuan Li
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Chuanshan Xu
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.
| | - Wei Deng
- School of Biomedical Engineering, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| | - Wenjie Chen
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.
| |
Collapse
|
5
|
Bahardoust M, Mousavi S, Moezi ZD, Yarali M, Tayebi A, Olamaeian F, Tizmaghz A. Effect of Metformin Use on Survival and Recurrence Rate of Gastric Cancer After Gastrectomy in Diabetic Patients: A Systematic Review and Meta-analysis of Observational Studies. J Gastrointest Cancer 2024; 55:65-76. [PMID: 37526857 DOI: 10.1007/s12029-023-00955-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2023] [Indexed: 08/02/2023]
Abstract
BACKGROUND Gastric cancer (GC) is one of the most common worldwide cancers and causes of death. Various studies have investigated the effect of metformin on overall survival (OS), cancer-specific survival (CSS), recurrence-free survival (RFS), and recurrence rate in diabetic patients after gastrectomy, and their results have been contradictory. This meta-analysis aimed to evaluate the effect of metformin use compared to sulfonylurea compounds with OS, CSS, RFS, and recurrence rate after gastrectomy in diabetic patients. METHODS We reviewed the Scopus, Google Scholar, PubMed, Web of Science, and Embassy databases until September 2022 based on appropriate MESH terms. All observational studies that evaluated the effect of metformin on survival in diabetic patients who underwent surgery for GC were included. The hazard ratio (HR) with a 95% confidence interval was used to estimate the effect size. The Egger test was used to evaluate publication bias. RESULTS Overall, nine studies, including 245,387 GC patients who underwent surgery, were included. The use of metformin significantly increased the OS rate (HR: 0.81, 95% CI: 0.78, 0.86, P: 0.001, I2: 4.5%), CSS rate (HR: 0.72, 95% CI: 0.63, 0.81, P: 0.011, I2 = 0%), and RFS rate (HR: 719, 95% CI: 0.524, 0.986, P: 0.001) and decreased the recurrence rate after gastrectomy (HR: 0.83, 95% CI: 0.77, 0.87, P: 0.001, I2: 0%). The use of metformin was significantly associated with a greater increase in OS and CSS rate and a greater decrease in recurrence rate in the Asian population than in the Western population. CONCLUSION The use of metformin in diabetic patients with GC can be associated with improved OS, CSS, RFS, and reduced recurrence rate after gastrectomy, especially in the Asian population.
Collapse
Affiliation(s)
- Mansour Bahardoust
- Department of Epidemiology, School of Public Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Safa Mousavi
- Department of Public Health, College of Health and Human Services, California State University, Fresno, CA, USA
| | - Zahra Deylami Moezi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Yarali
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Tayebi
- Department of General Surgery, School of Medicine, Firoozabadi Clinical Research Development Unit (FACRDU), Iran University of Medical Sciences, Tehran, Iran.
| | - Faranak Olamaeian
- Department of General Surgery, School of Medicine, Firoozabadi Clinical Research Development Unit (FACRDU), Iran University of Medical Sciences, Tehran, Iran
| | - Adnan Tizmaghz
- Department of General Surgery, School of Medicine, Firoozabadi Clinical Research Development Unit (FACRDU), Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Kalajahi HG, Yari A, Amini M, Catal T, Ahmadpour Youshanlui M, Pourbagherian O, Zhmurov CS, Mokhtarzadeh A. Therapeutic effect of microRNA-21 on differentially expressed hub genes in gastric cancer based on systems biology. Sci Rep 2023; 13:21906. [PMID: 38081950 PMCID: PMC10713559 DOI: 10.1038/s41598-023-49225-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
Gastric cancer (GC) is a leading cause of mortality for many people. Cancer's initiating factors are poorly understood. miR-21 has a crucial function in several malignancies, particularly GC. Furthermore, it has been shown that miR-21 is critical for the emergence and advancement of GC. This work intends to identify new genes which expression is associated with the activity of mir-21 in GC and to investigate the effect of downregulation of mir-21 on these genes and gastric tumorigenesis. We utilized the gene expression profiles of GCs from an Array database (GSE13911) from the Gene Expression Omnibus (GEO) dataset to find differentially expressed genes (DEGs) between control and gastric cancer groups. Using weighted gene correlation network analysis (WGCNA) in R, the Gene co-expression network was reconstructed. The microRNA-mRNA network was then reconstructed using the miRWalk database, and by investigating the microRNA-mRNA network, the genes that have an association with mir-21 were found. To implement the functional investigation, MKN and AGS cell lines were transfected with anti-miR-21 next. Subsequently, MTT proliferation was utilized to assess the cell's vitality. qRT-PCR was then used to evaluate the anticipated levels of gene expression in both GC cell lines. This study discovered and predicted CCL28, NR3C2, and SNYPO2 as the targets of miR-21 (GC), which are downregulated through gastric tumorigenesis, showing great potential as therapeutic and diagnostic targets. The suppression of miR-21 in gastric GC cells led to the inhibition of cell proliferation and decreased expression of CCL28, NR3C2, and SNYPO2 genes. This study established that miR-21, via downregulating these genes, contributes significantly to the development of GC. In addition, systems biology techniques identified CCL28, NR3C2, and SNYPO2 genes as possible GC surveillance and therapy components.
Collapse
Affiliation(s)
- Hesam Ghafouri Kalajahi
- Department of Molecular Biology and Genetics, Uskudar University, Uskudar, 34662, Istanbul, Turkey
| | - AmirHossein Yari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tunc Catal
- Department of Molecular Biology and Genetics, Uskudar University, Uskudar, 34662, Istanbul, Turkey
| | | | - Omid Pourbagherian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Cigdem Sezer Zhmurov
- Department of Molecular Biology and Genetics, Uskudar University, Uskudar, 34662, Istanbul, Turkey.
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
Joliat GR, Gaspar-Figueiredo S, Labgaa I, Vrochides D, Perinel J, Adham M, Demartines N, Schäfer M. Impact of preoperative statin medication on long-term outcomes after pancreatoduodenectomy for ductal adenocarcinoma: an international multicentric cross-sectional study. J Cancer Res Clin Oncol 2023; 149:17007-17014. [PMID: 37740766 PMCID: PMC10657279 DOI: 10.1007/s00432-023-05426-2] [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: 08/04/2023] [Accepted: 09/13/2023] [Indexed: 09/25/2023]
Abstract
PURPOSE Statin treatment has been shown in certain population studies and meta-analyses to improve survival of patients with pancreatic ductal adenocarcinoma (PDAC). This study assessed if patients with statin treatment had better overall survival (OS) and disease-free survival (DFS) after upfront pancreatoduodenectomy for PDAC. METHODS Consecutive PDAC patients were retrospectively collected from three centers in Europe and USA (study period: 2000-2017). Adult patients who underwent upfront pancreatoduodenectomy and survived the first 90 postoperative days were included. Patients with metastasis at diagnosis or with macroscopic incomplete resection were excluded. Patients were considered under statin if started at least one month before pancreatoduodenectomy. Survival rates were calculated using Kaplan-Meier method and compared with log-rank test. RESULTS A total of 496 patients were included. Median age was 67 years (IQR 59-75), 48% (n = 236) were women, and 141 patients (28%) received statin treatment already preoperatively. Patients with and without statin treatment were comparable in terms of demographics and pre-/intraoperative characteristics, except for age and pre-existing diabetes. Median OS and DFS were similar in patients with and without statin treatment (OS: 29, 95% CI 22-36 vs. 27 months, 95% CI 22-32, p = 0.370, DFS: 18, 95% CI 14-22 vs. 16 months, 95% CI 14-18, p = 0.430). On multivariable Cox regression, lymph node involvement (HR 1.9, 95% CI 1.6-2.2, p < 0.001), tumor differentiation (HR 1.3, 95% CI 1.1-1.6, p = 0.003), and postoperative chemotherapy (HR 0.5, 95% CI 0.4-0.7, p < 0.001) were predictors of OS, whereas statin treatment was not a prognostic factor (HR 0.9, 95% CI 0.7-1.2, p = 0.376). CONCLUSION In this international cohort of PDAC patients, statin treatment did not influence survival after upfront pancreatoduodenectomy. Nodal involvement, tumor differentiation, and postoperative chemotherapy were independent predictors of OS.
Collapse
Affiliation(s)
- Gaëtan-Romain Joliat
- Department of Visceral Surgery, Lausanne University Hospital CHUV, University of Lausanne (UNIL), Rue du Bugnon 46, 1011, Lausanne, Switzerland.
- Graduate School of Health Sciences, University of Bern, Bern, Switzerland.
| | - Sérgio Gaspar-Figueiredo
- Department of Visceral Surgery, Lausanne University Hospital CHUV, University of Lausanne (UNIL), Rue du Bugnon 46, 1011, Lausanne, Switzerland
| | - Ismail Labgaa
- Department of Visceral Surgery, Lausanne University Hospital CHUV, University of Lausanne (UNIL), Rue du Bugnon 46, 1011, Lausanne, Switzerland
| | - Dionisios Vrochides
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Carolinas Medical Center, Charlotte, USA
| | - Julie Perinel
- Department of Surgery, Edouard Herriot Hospital, Lyon, France
| | - Mustapha Adham
- Department of Surgery, Edouard Herriot Hospital, Lyon, France
| | - Nicolas Demartines
- Department of Visceral Surgery, Lausanne University Hospital CHUV, University of Lausanne (UNIL), Rue du Bugnon 46, 1011, Lausanne, Switzerland
| | - Markus Schäfer
- Department of Visceral Surgery, Lausanne University Hospital CHUV, University of Lausanne (UNIL), Rue du Bugnon 46, 1011, Lausanne, Switzerland
| |
Collapse
|
8
|
Podoltsev NA, Wang R, Shallis RM, Stempel JM, Di M, Neparidze N, Zeidan AM, Huntington SF, Giri S, Hull SC, Gore SD, Ma X. Statin use, survival and incidence of thrombosis among older patients with polycythemia vera and essential thrombocythemia. Cancer Med 2023; 12:18889-18900. [PMID: 37702132 PMCID: PMC10557879 DOI: 10.1002/cam4.6528] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/22/2023] [Accepted: 08/31/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Polycythemia vera (PV) and essential thrombocythemia (ET) are linked to increased risk of cardiovascular morbidity and mortality. In addition to the reduction in of arterial thrombotic events, statins may prevent venous thrombosis including among patients with cancer. As previous registry- and claims-based studies revealed that the use of statins may improve the survival of patients with various malignancies we evaluated their impact on outcomes of older adults with PV and ET. METHODS We identified 4010 older adults (aged 66-99 years at diagnosis) with PV (n = 1809) and ET (n = 2201) in a population-based cohort study using the Surveillance, Epidemiology, and End Results-Medicare database with median follow-up of 3.92 (interquartile range: 2.58-5.75) years. Propensity score matching (PSM) and inverse probability of treatment weighting (IPTW) approaches were utilized to assess potential association between statins and overall survival. Multivariable competing risk models with death as a competing risk were used to evaluate possible relationship between statins and the incidence of thrombosis. RESULTS 55.8% of the patients used statins within the first year after PV/ET diagnosis, and statin use was associated with a 22% reduction in all-cause mortality (PSM: hazard ratio [HR] = 0.78, 95% confidence interval [CI]: 0.63-0.98, p = 0.03; IPTW: HR = 0.79, 95% CI: 0.64-0.97, p = 0.03). Statins also reduced the risk of thrombosis in this patient population (PSM: HR = 0.63, 95% CI: 0.51-0.78, p < 0.01; IPTW: HR = 0.57, 95% CI: 0.49-0.66, p < 0.01) as well as in PV and ET subgroups. CONCLUSIONS These findings suggest that it may be important to incorporate statins into the therapeutic strategy for older adults with PV and ET.
Collapse
Affiliation(s)
- Nikolai A. Podoltsev
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
| | - Rong Wang
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
- Department of Chronic Disease EpidemiologyYale School of Public HealthNew HavenConnecticutUSA
| | - Rory M. Shallis
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
| | - Jessica M. Stempel
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
| | - Mengyang Di
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
| | - Natalia Neparidze
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
| | - Amer M. Zeidan
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
| | - Scott F. Huntington
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
| | - Smith Giri
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Present address:
Division of Hematology and OncologyUniversity of Alabama School of MedicineBirminghamAlabamaUSA
| | - Sarah C. Hull
- Section of Cardiology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Steven D. Gore
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Present address:
Investigational Drug Branch, Cancer Therapy Evaluation ProgramNational Cancer InstituteBethesdaMarylandUSA
| | - Xiaomei Ma
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
- Department of Chronic Disease EpidemiologyYale School of Public HealthNew HavenConnecticutUSA
| |
Collapse
|
9
|
Huang Z, Zhou J, Chen L, Zhang Y. Metabolic Syndrome and Clinical Outcomes of Patients with Gastric Cancer: A Meta-Analysis. Horm Metab Res 2023; 55:333-342. [PMID: 37011889 DOI: 10.1055/a-2038-5830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Metabolic syndrome (MetS) is suggested to participate in the pathogenesis and progress of some cancers via inducing low-grade systemic inflammation. However, the influence of MetS on patients with gastric cancer (GC) remains not fully determined. A systematic review and meta-analysis was therefore performed to evaluate the influence of MetS on clinical outcomes of patients with GC. A search of PubMed, Embase, Web of Science, Wanfang, and CNKI retrieved relevant cohort studies from the inception of the databases to October 11, 2022. We pooled the results using a random-effects model that incorporates heterogeneity. In the meta-analysis, 6649 patients with GC were included, and all of them received gastrectomy. A total of 1248 (18.8%) patients had MetS at baseline. Pooled results showed that MetS was associated with higher risks of postoperative complications [risk ratio (RR): 2.41, 95% confidence interval (CI): 1.85 to 3.14, p<0.001; I2=55%], overall mortality (RR: 1.73, 95% CI: 1.85 to 3.14, p<0.001; I2=77%), and recurrence of GC (RR: 2.00, 95% CI: 1.10 to 3.63, p=0.02; I2=39%). Subgroup analyses showed similar results in prospective and retrospective cohort studies and in studies with MetS diagnosed with the Chinese Diabetes Society criteria and the National Cholesterol Education Program Adult Treatment Panel III criteria (p for subgroup difference all>0.05). In patients with GC after gastrectomy, MetS may be a predictor of high incidence of postoperative complications, cancer recurrence, and overall mortality.
Collapse
Affiliation(s)
- Zhiyang Huang
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, China
| | - Jianwei Zhou
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, China
| | - Long Chen
- Center for Rehabilitation Medicine and Department of Anesthesiology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Yibing Zhang
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|