1
|
Micucci M, Xiang BZ, Ting CM, Kwan HY, Mari M, Retini M, Burattini S, Osman R, Okeke UJ, Abdullah FO, Gianfanti F, Battistelli M. Matching traditional Chinese medicine and western medicine-based research: Advanced nutraceutical development for proactive gastric cancer prevention. World J Gastrointest Oncol 2024; 16:3798-3819. [PMID: 39350981 PMCID: PMC11438774 DOI: 10.4251/wjgo.v16.i9.3798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/26/2024] [Accepted: 07/24/2024] [Indexed: 09/09/2024] Open
Abstract
Gastric cancer (GC), the third leading cause of cancer-related death globally, is complex and heterogeneous. This review explores multidisciplinary investigations of traditional Chinese medicine (TCM) combined with Western medical practices, emphasizing the development of nutraceuticals for cancer prevention. Using advanced analytical chemistry and food chemistry techniques, this study investigated how TCM components may be optimized for nutraceutical development. Focusing on molecular interactions with GC pathways, particularly the NF-κB, PI3K/Akt, and Wnt/β-catenin pathways, we examined the effects of TCM polyherbal formulas, extracts, and isolated compounds. These agents modulate apoptosis and cellular proliferation, underscoring their potential in preventive strategies. The convergence of nutraceutical and medicine food homology studies highlights a significant shift towards integrating TCM-derived compounds in a preventive health framework. This approach aims not only to enhance efficacy and reduce side effects but also to champion a preventive paradigm using personalized medicine to advance proactive health maintenance and disease prevention. The combination of TCM and western medical practices offers promising avenues for future research and practical applications in GC prevention.
Collapse
Affiliation(s)
- Matteo Micucci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino 61029, Italy
| | - Bian-Zhao Xiang
- Hong Kong Chinese Medicine Clinical Study Centre, Chinese EQUATOR Centre, School of Chinese Medicine, Chinese Clinical Trial Registry (Hong Kong), Hong Kong Baptist University, Hong Kong 999077, China
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong 999077, China
| | - Chen-Min Ting
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China
| | - Hiu-Yee Kwan
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China
| | - Michele Mari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino 61029, Italy
| | - Michele Retini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino 61029, Italy
| | - Sabrina Burattini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino 61029, Italy
| | - Riham Osman
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino 61029, Italy
| | - Udodinma Jude Okeke
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino 61029, Italy
| | - Fuad Othman Abdullah
- Department of Chemistry, College of Science, Salahaddin University-Erbil, Erbil 44001, Iraq
- Department of Pharmacognosy, Faculty Pharmacy, Tishk International University, Erbil 44001, Iraq
| | - Federico Gianfanti
- Institute of Oncology Research, Bellinzona CH6500, Switzerland
- Università della Svizzera Italiana, Lugano CH6900, Switzerland
| | - Michela Battistelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino 61029, Italy
| |
Collapse
|
2
|
Cook SR, Hugen S, Hayward JJ, Famula TR, Belanger JM, McNiel E, Fieten H, Oberbauer AM, Leegwater PA, Ostrander EA, Mandigers PJ, Evans JM. Genomic analyses identify 15 susceptibility loci and reveal HDAC2, SOX2-OT, and IGF2BP2 in a naturally-occurring canine model of gastric cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.14.604426. [PMID: 39372775 PMCID: PMC11451740 DOI: 10.1101/2024.08.14.604426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Gastric cancer (GC) is the fifth most common human cancer worldwide, but the genetic etiology is largely unknown. We performed a Bayesian genome-wide association study and selection analyses in a naturally-occurring canine model of GC, the Belgian Tervuren and Sheepdog breeds, to elucidate underlying genetic risk factors. We identified 15 loci with over 90% predictive accuracy for the GC phenotype. Variant filtering revealed germline putative regulatory variants for the EPAS1 (HIF2A) and PTEN genes and a coding variant in CD101. Although closely related to Tervuren and Sheepdogs, Belgian Malinois rarely develop GC. Across-breed analyses uncovered protective haplotypes under selection in Malinois at SOX2-OT and IGF2BP2. Among Tervuren and Sheepdogs, HDAC2 putative regulatory variants were present at comparatively high frequency and were associated with GC. Here, we describe a complex genetic architecture governing GC in a dog model, including genes such as PDZRN3, that have not been associated with human GC.
Collapse
Affiliation(s)
- Shawna R. Cook
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Sanne Hugen
- Expertisecentre of Genetics, Department of Clinical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jessica J. Hayward
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Thomas R. Famula
- Department of Animal Science, University of California, Davis, CA, USA
| | | | - Elizabeth McNiel
- Cummings School of Veterinary Medicine, Tufts University, Grafton, Massachusetts, USA
| | - Hille Fieten
- Expertisecentre of Genetics, Department of Clinical Sciences, Utrecht University, Utrecht, The Netherlands
| | | | - Peter A.J. Leegwater
- Expertisecentre of Genetics, Department of Clinical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Elaine A. Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Center, National Institutes of Health, Bethesda, MD, USA
| | - Paul J.J. Mandigers
- Expertisecentre of Genetics, Department of Clinical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jacquelyn M. Evans
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| |
Collapse
|
3
|
Fernández Aceñero MJ, Díaz del Arco C. Hereditary Gastrointestinal Tumor Syndromes: When Risk Comes with Your Genes. Curr Issues Mol Biol 2024; 46:6440-6471. [PMID: 39057027 PMCID: PMC11275188 DOI: 10.3390/cimb46070385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Despite recent campaigns for screening and the latest advances in cancer therapy and molecular biology, gastrointestinal (GI) neoplasms remain among the most frequent and lethal human tumors. Most GI neoplasms are sporadic, but there are some well-known familial syndromes associated with a significant risk of developing both benign and malignant GI tumors. Although some of these entities were described more than a century ago based on clinical grounds, the increasing molecular information obtained with high-throughput techniques has shed light on the pathogenesis of several of them. The vast amount of information gained from next-generation sequencing has led to the identification of some high-risk genetic variants, although others remain to be discovered. The opportunity for genetic assessment and counseling in these families has dramatically changed the management of these syndromes, though it has also resulted in significant psychological distress for the affected patients, especially those with indeterminate variants. Herein, we aim to summarize the most relevant hereditary cancer syndromes involving the stomach and colon, with an emphasis on new molecular findings, novel entities, and recent changes in the management of these patients.
Collapse
Affiliation(s)
- María Jesús Fernández Aceñero
- Department of Legal Medicine, Psychiatry and Pathology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Pathology, Hospital Clínico San Carlos, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Cristina Díaz del Arco
- Department of Legal Medicine, Psychiatry and Pathology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Pathology, Hospital Clínico San Carlos, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| |
Collapse
|
4
|
Carballal S, Balaguer F, Bujanda L, Capellá G, González Santiago S, Jover R, Moreira L, Pineda M, Ruiz-Ponte C, Sánchez Heras AB, Serrano Blanch R, Soto JL, Vidal Tocino R, Cubiella J. Use of multi-gene panels in patients at high risk of hereditary digestive cancer: position statement of AEG, SEOM, AEGH and IMPaCT-GENÓMICA consortium. GASTROENTEROLOGIA Y HEPATOLOGIA 2024; 47:293-318. [PMID: 37315767 DOI: 10.1016/j.gastrohep.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/04/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
This position statement, sponsored by the Asociación Española de Gastroenterología, the Sociedad Española de Oncología Médica, the Asociación Española de Genética Humana and the IMPaCT-Genómica Consortium aims to establish recommendations for use of multi-gene panel testing in patients at high risk of hereditary gastrointestinal and pancreatic cancer. To rate the quality of the evidence and the levels of recommendation, we used the methodology based on the GRADE system (Grading of Recommendations Assessment, Development and Evaluation). We reached a consensus among experts using a Delphi method. The document includes recommendations on clinical scenarios where multi-gene panel testing is recommended in colorectal cancer, polyposis syndromes, gastric and pancreatic cancer, as well as the genes to be considered in each clinical scenario. Recommendations on the evaluation of mosaicisms, counseling strategies in the absence of an index subject and, finally, constitutional analysis after identification of pathogenic tumor variants are also made.
Collapse
Affiliation(s)
- Sabela Carballal
- Servicio de Gastroenterología, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, España.
| | - Francesc Balaguer
- Servicio de Gastroenterología, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, España
| | - Luis Bujanda
- Servicio de Aparato Digestivo, Hospital Universitario Donostia, Instituto Biodonostia. Universidad del País Vasco (UPV/EHU), CIBEREHD, San Sebastián, Guipúzcoa, España
| | - Gabriel Capellá
- Programa de Cáncer Hereditario, Instituto Catalán de Oncología, Programa ONCOBELL, IDIBELL, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), L'Hospitalet de Llobregat, Barcelona, España
| | | | - Rodrigo Jover
- Servicio de Medicina Digestiva, Hospital General Universitario Dr. Balmis, Instituto de Investigación Sanitaria de Alicante (ISABIAL), Departamento de Medicina Clínica, Universidad Miguel Hernández, Alicante, España
| | - Leticia Moreira
- Servicio de Gastroenterología, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, España
| | - Marta Pineda
- Programa de Cáncer Hereditario, Instituto Catalán de Oncología, Programa ONCOBELL, IDIBELL, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), L'Hospitalet de Llobregat, Barcelona, España
| | - Clara Ruiz-Ponte
- Fundación Pública Galega de Medicina Xenómica (SERGAS), Instituto de Investigación Sanitaria de Santiago (IDIS), Grupo de Medicina Xenomica (USC), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERer), Santiago de Compostela, La Coruña, España
| | - Ana Beatriz Sánchez Heras
- Unidad de Consejo Genético en Cáncer, Servicio de Oncología Médica, Hospital General Universitario de Elche, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Elche, Alicante, España
| | - Raquel Serrano Blanch
- Unidad de Consejo Genético en Cáncer, Unidad de Gestión Clínica de Oncología Médica, H.U. Reina Sofía de Córdoba. Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), CIBERONC, Universidad de Córdoba (UCO), Córdoba, España
| | - José Luis Soto
- Unidad de Genética Molecular, Hospital General Universitario de Elche, FISABIO, Elche, Alicante, España
| | - Rosario Vidal Tocino
- Servicio de Oncología Médica, Complejo Asistencial Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, España
| | - Joaquín Cubiella
- Servicio de Aparato Digestivo, Hospital Universitario de Ourense, Grupo de Investigación en Oncología Digestiva-Ourense (GIODO), CIBEREHD, Ourense, España.
| |
Collapse
|
5
|
São José C, Garcia-Pelaez J, Ferreira M, Arrieta O, André A, Martins N, Solís S, Martínez-Benítez B, Ordóñez-Sánchez ML, Rodríguez-Torres M, Sommer AK, Te Paske IBAW, Caldas C, Tischkowitz M, Tusié MT, Hoogerbrugge N, Demidov G, de Voer RM, Laurie S, Oliveira C. Combined loss of CDH1 and downstream regulatory sequences drive early-onset diffuse gastric cancer and increase penetrance of hereditary diffuse gastric cancer. Gastric Cancer 2023; 26:653-666. [PMID: 37249750 PMCID: PMC10361908 DOI: 10.1007/s10120-023-01395-0] [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: 03/08/2023] [Accepted: 04/30/2023] [Indexed: 05/31/2023]
Abstract
BACKGROUND Germline CDH1 pathogenic or likely pathogenic variants cause hereditary diffuse gastric cancer (HDGC). Once a genetic cause is identified, stomachs' and breasts' surveillance and/or prophylactic surgery is offered to asymptomatic CDH1 carriers, which is life-saving. Herein, we characterized an inherited mechanism responsible for extremely early-onset gastric cancer and atypical HDGC high penetrance. METHODS Whole-exome sequencing (WES) re-analysis was performed in an unsolved HDGC family. Accessible chromatin and CDH1 promoter interactors were evaluated in normal stomach by ATAC-seq and 4C-seq, and functional analysis was performed using CRISPR-Cas9, RNA-seq and pathway analysis. RESULTS We identified a germline heterozygous 23 Kb CDH1-TANGO6 deletion in a family with eight diffuse gastric cancers, six before age 30. Atypical HDGC high penetrance and young cancer-onset argued towards a role for the deleted region downstream of CDH1, which we proved to present accessible chromatin, and CDH1 promoter interactors in normal stomach. CRISPR-Cas9 edited cells mimicking the CDH1-TANGO6 deletion display the strongest CDH1 mRNA downregulation, more impacted adhesion-associated, type-I interferon immune-associated and oncogenic signalling pathways, compared to wild-type or CDH1-deleted cells. This finding solved an 18-year family odyssey and engaged carrier family members in a cancer prevention pathway of care. CONCLUSION In this work, we demonstrated that regulatory elements lying down-stream of CDH1 are part of a chromatin network that control CDH1 expression and influence cell transcriptome and associated signalling pathways, likely explaining high disease penetrance and very young cancer-onset. This study highlights the importance of incorporating scientific-technological updates and clinical guidelines in routine diagnosis, given their impact in timely genetic diagnosis and disease prevention.
Collapse
Affiliation(s)
- Celina São José
- i3S-Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- IPATIMUP-Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- Doctoral Programme in Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
| | - José Garcia-Pelaez
- i3S-Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- IPATIMUP-Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- Doctoral Programme in Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Marta Ferreira
- i3S-Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- IPATIMUP-Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- Department Computer Science Faculty of Science, University of Porto, Porto, Portugal
| | - Oscar Arrieta
- Thoracic Oncology Unit, Department of Thoracic Oncology, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Ana André
- i3S-Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- IPATIMUP-Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Nelson Martins
- i3S-Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- IPATIMUP-Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- Master Programme in Molecular Medicine and Oncology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Samantha Solís
- INCMNSZ/Instituto de Investigaciones Biomédicas, Unidad de Biología Molecular y Medicina Genómica Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, UNAM Mexico City, Mexico
| | - Braulio Martínez-Benítez
- Pathology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, INCMNSZ Mexico City, Mexico
| | - María Luisa Ordóñez-Sánchez
- INCMNSZ/Instituto de Investigaciones Biomédicas, Unidad de Biología Molecular y Medicina Genómica Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, UNAM Mexico City, Mexico
| | - Maribel Rodríguez-Torres
- INCMNSZ/Instituto de Investigaciones Biomédicas, Unidad de Biología Molecular y Medicina Genómica Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, UNAM Mexico City, Mexico
| | - Anna K Sommer
- Institute of Human Genetics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Iris B A W Te Paske
- Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
- Cambridge Experimental Cancer Medicine Centre (ECMC), CRUK Cambridge Centre, NIHR Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Marc Tischkowitz
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Maria Teresa Tusié
- INCMNSZ/Instituto de Investigaciones Biomédicas, Unidad de Biología Molecular y Medicina Genómica Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, UNAM Mexico City, Mexico
| | - Nicoline Hoogerbrugge
- Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - German Demidov
- Institute of Medical Genetics and Applied Genomics, Tübingen, Germany
| | - Richarda M de Voer
- Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Steve Laurie
- The Barcelona Institute of Science and Technology, CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona, Spain
| | - Carla Oliveira
- i3S-Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.
- IPATIMUP-Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal.
- FMUP-Faculty of Medicine of the University of Porto, Porto, Portugal.
| |
Collapse
|
6
|
Calvello M, Marabelli M, Gandini S, Marino E, Bernard L, Dal Molin M, Di Cola G, Zanzottera C, Corso G, Fazio N, Gervaso L, Fumagalli Romario U, Barberis M, Guerrieri-Gonzaga A, Bertario L, Serrano D, Bonanni B. Hereditary Gastric Cancer: Single-Gene or Multigene Panel Testing? A Mono-Institutional Experience. Genes (Basel) 2023; 14:genes14051077. [PMID: 37239438 DOI: 10.3390/genes14051077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Gastric cancer (GC) has long been a 'Cinderella' among hereditary cancers. Until recently, single-gene testing (SGT) was the only approach to identify high-risk individuals. With the spread of multigene panel testing (MGPT), a debate arose on the involvement of other genes, particularly those pertaining to homologous recombination (HR) repair. We report our mono-institutional experience in genetic counseling and SGT for 54 GC patients, with the detection of nine pathogenic variants (PVs) (9/54:16.7%). Seven out of fifty (14%) patients who underwent SGT for unknown mutations were carriers of a PV in CDH1 (n = 3), BRCA2 (n = 2), BRCA1 (n = 1), and MSH2 (n = 1), while one patient (2%) carried two variants of unknown significance (VUSs). CDH1 and MSH2 emerged as genes involved in early-onset diffuse and later-onset intestinal GCs, respectively. We additionally conducted MGPT on 37 patients, identifying five PVs (13.5%), including three (3/5:60%) in an HR gene (BRCA2, ATM, RAD51D) and at least one VUS in 13 patients (35.1%). Comparing PV carriers and non-carriers, we observed a statistically significant difference in PVs between patients with and without family history of GC (p-value: 0.045) or Lynch-related tumors (p-value: 0.036). Genetic counseling remains central to GC risk assessment. MGPT appeared advantageous in patients with unspecific phenotypes, although it led to challenging results.
Collapse
Affiliation(s)
- Mariarosaria Calvello
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Monica Marabelli
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Sara Gandini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Elena Marino
- Clinic Unit of Oncogenomics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Loris Bernard
- Clinic Unit of Oncogenomics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Matteo Dal Molin
- Clinic Unit of Oncogenomics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giulia Di Cola
- Clinic Unit of Oncogenomics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Cristina Zanzottera
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giovanni Corso
- Division of Breast Surgery, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
- European Cancer Prevention Organization (ECP), 20122 Milan, Italy
| | - Nicola Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Lorenzo Gervaso
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Molecular Medicine Program, University of Pavia, 27100 Pavia, Italy
| | | | - Massimo Barberis
- Clinic Unit of Oncogenomics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Aliana Guerrieri-Gonzaga
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Lucio Bertario
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Davide Serrano
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| |
Collapse
|
7
|
Zou W, Zhang Q, Sun R, Li X, He S. Study on TFF1 and PALB2 gene variants associated with gastric carcinoma risk in the Chinese Han population. Cancer Epidemiol 2023; 83:102333. [PMID: 36758349 DOI: 10.1016/j.canep.2023.102333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 01/09/2023] [Accepted: 01/14/2023] [Indexed: 02/10/2023]
Abstract
OBJECTIVE Gastric carcinoma (GC) has received extensive attention due to its complex pathogenesis. Studies have shown that the expression of Trefoil factor 1 (TFF1) and Partner and localiser of BRCA2 (PALB2) genes promotes the occurrence of GC. Therefore, we investigated whether TFF1 and PALB2 gene polymorphisms are associated with GC risk in the Chinese Han population. METHODS A total of 509 GC cases and 505 controls were recruited, and single nucleotide polymorphisms (SNPs) of TFF1 and PALB2 in these subjects were genotyped. The association between each candidate polymorphism and GC risk was assessed by calculating odds ratios (ORs) and 95% confidence intervals (CIs). The visualization of gene-gene interactions and functional enrichment analysis were then performed using Cytoscape software and the R package "cluster profile". RESULTS The TFF1 rs2156310 polymorphism significantly reduced the predisposition to GC in people under 60 years of age (AA vs. AG - GG, OR = 0.58, 95% CI = 0.35-0.97, p = 0.036). The gender-stratified analysis found that PALB2 rs513313 was significantly associated with the risk of GC in males (CT vs. TT, OR = 1.51, 95% CI = 1.06-2.15, p = 0.022). Besides, PALB2 rs249954 significantly reduced the susceptibility to GC in females (AA vs GG, OR = 0.42, 95% CI = 0.19-0.94, p = 0.034). CONCLUSION Our results revealed that TFF1 and PALB2 gene polymorphisms were correlated with the genetic susceptibility to GC, providing certain data support for researchers to further study the mechanism of GC.
Collapse
Affiliation(s)
- Wenjing Zou
- Department of Digestive Internal Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Department of Digestive Internal Medicine, Xi'an Fifth Hospital, Xi'an 710082, China
| | - Qian Zhang
- Department of First Internal Medicine, Shaanxi Province Cancer Hospital, Xi'an 710061, China
| | - Ruifang Sun
- Department of Pathology, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Xu Li
- Department of Sixth Internal Medicine, Shaanxi Province Cancer Hospital, Xi'an 710061, China.
| | - Shuixiang He
- Department of Digestive Internal Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
8
|
Vetti HH, Blanco I, Menko FH. European collaboration on genetic tumour risk syndromes. Eur J Med Genet 2023; 66:104691. [PMID: 36581153 DOI: 10.1016/j.ejmg.2022.104691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Hildegunn H Vetti
- Western Norway Familial Cancer Center, Haukeland University Hospital, Bergen, Norway and VID Specialized University, Faculty of Health Studies, Bergen, Norway.
| | - Ignacio Blanco
- Clinical Genetics Department, Hospital Germans Trias I Pujol, Barcelona, Spain
| | - Fred H Menko
- Family Cancer Clinic, Antoni van Leeuwenhoek Hospital/ The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
9
|
Badie A, Gaiddon C, Mellitzer G. Histone Deacetylase Functions in Gastric Cancer: Therapeutic Target? Cancers (Basel) 2022; 14:5472. [PMID: 36358890 PMCID: PMC9659209 DOI: 10.3390/cancers14215472] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 09/05/2023] Open
Abstract
Gastric cancer (GC) is one of the most aggressive cancers. Therapeutic treatments are based on surgery combined with chemotherapy using a combination of platinum-based agents. However, at metastatic stages of the disease, survival is extremely low due to late diagnosis and resistance mechanisms to chemotherapies. The development of new classifications has not yet identified new prognostic markers for clinical use. The studies of epigenetic processes highlighted the implication of histone acetylation status, regulated by histone acetyltransferases (HATs) and by histone deacetylases (HDACs), in cancer development. In this way, inhibitors of HDACs (HDACis) have been developed and some of them have already been clinically approved to treat T-cell lymphoma and multiple myeloma. In this review, we summarize the regulations and functions of eighteen HDACs in GC, describing their known targets, involved cellular processes, associated clinicopathological features, and impact on survival of patients. Additionally, we resume the in vitro, pre-clinical, and clinical trials of four HDACis approved by Food and Drug Administration (FDA) in cancers in the context of GC.
Collapse
Affiliation(s)
| | | | - Georg Mellitzer
- Laboratoire Streinth, Université de Strasbourg, Inserm UMR_S 1113 IRFAC, 67200 Strasbourg, France
| |
Collapse
|
10
|
Lordick F, Carneiro F, Cascinu S, Fleitas T, Haustermans K, Piessen G, Vogel A, Smyth EC. Gastric cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2022; 33:1005-1020. [PMID: 35914639 DOI: 10.1016/j.annonc.2022.07.004] [Citation(s) in RCA: 495] [Impact Index Per Article: 165.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 02/08/2023] Open
Affiliation(s)
- F Lordick
- Department of Medicine II (Oncology, Gastroenterology, Hepatology, Pulmonology and Infectious Diseases), University Cancer Center Leipzig (UCCL), University Medical Center, Leipzig, Germany
| | - F Carneiro
- Department of Pathology, Centro Hospitalar Universitário de São João (CHUSJ), Porto; Faculty of Medicine, University of Porto (FMUP), Porto; Instituto de Investigação e Inovação em Saúde (i3S)/Institute of Molecular Pathology and Immunology, University of Porto (Ipatimup), Porto, Portugal
| | - S Cascinu
- Department of Medical Oncology, Comprehensive Cancer Center, Università Vita-Salute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - T Fleitas
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - K Haustermans
- Department of Radiation Oncology, University Hospital Leuven, Leuven, Belgium
| | - G Piessen
- University of Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille;; CNRS, UMR9020, Lille; Inserm, U1277, Lille; CHU Lille, Department of Digestive and Oncological Surgery, Lille, France
| | - A Vogel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - E C Smyth
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|