1
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Current chemoprevention approaches in Lynch syndrome and Familial adenomatous polyposis: a global clinical practice survey

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          International chemoprevention preferences and approaches in Lynch syndrome (LS) and APC-associated polyposis, including Familial adenomatous polyposis (FAP) and attenuated FAP (AFAP) have not been previously explored.

          Aim

          To describe current chemoprevention strategies for patients with LS or FAP/AFAP (referred to collectively as FAP) practiced by members of four international hereditary cancer societies through administration of a survey.

          Results

          Ninety-six participants across four hereditary gastrointestinal cancer societies responded to the survey. Most respondents (91%, 87/96) completed information regarding their demographics and practice characteristics relating to hereditary gastrointestinal cancer and chemoprevention clinical practices. Sixty-nine percent (60/87) of respondents offer chemoprevention for FAP and/or LS as a part of their practice. Of the 75% (72/96) of survey respondents who were eligible to answer practice-based clinical vignettes based off of their responses to ten barrier questions regarding chemoprevention, 88% (63/72) of those participants completed at least one case vignette question to further characterize chemoprevention practices in FAP and/or LS. In FAP, 51% (32/63) would offer chemoprevention for rectal polyposis, with sulindac - 300 mg (18%, 10/56) and aspirin (16%, 9/56) being the most frequently selected options. In LS, 93% (55/59) of professionals discuss chemoprevention and 59% (35/59) frequently recommend chemoprevention. Close to half of the respondents (47%, 26/55) would recommend beginning aspirin at time of commencement of the patient’s first screening colonoscopy (usually at age 25yrs). Ninety-four percent (47/50) of respondents would consider a patient’s diagnosis of LS as an influential factor for aspirin use. There was no consensus on the dose of aspirin (≤100 mg, >100 mg - 325 mg or 600 mg) to offer patients with LS and there was no agreement on how other factors, such as BMI, hypertension, family history of colorectal cancer, and family history of heart disease, would affect the recommendation for aspirin use. Possible harm among older patients (>70 years) was identified as the most common reason to discourage aspirin use.

          Conclusion

          Although chemoprevention is widely discussed and offered to patients with FAP and LS by an international group of hereditary gastrointestinal cancer experts, there is significant heterogeneity in how it is applied in clinical practice.

          Related collections

          Most cited references33

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          A simple practice guide for dose conversion between animals and human

          Understanding the concept of extrapolation of dose between species is important for pharmaceutical researchers when initiating new animal or human experiments. Interspecies allometric scaling for dose conversion from animal to human studies is one of the most controversial areas in clinical pharmacology. Allometric approach considers the differences in body surface area, which is associated with animal weight while extrapolating the doses of therapeutic agents among the species. This review provides basic information about translation of doses between species and estimation of starting dose for clinical trials using allometric scaling. The method of calculation of injection volume for parenteral formulation based on human equivalent dose is also briefed.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            ACG clinical guideline: Genetic testing and management of hereditary gastrointestinal cancer syndromes.

            This guideline presents recommendations for the management of patients with hereditary gastrointestinal cancer syndromes. The initial assessment is the collection of a family history of cancers and premalignant gastrointestinal conditions and should provide enough information to develop a preliminary determination of the risk of a familial predisposition to cancer. Age at diagnosis and lineage (maternal and/or paternal) should be documented for all diagnoses, especially in first- and second-degree relatives. When indicated, genetic testing for a germline mutation should be done on the most informative candidate(s) identified through the family history evaluation and/or tumor analysis to confirm a diagnosis and allow for predictive testing of at-risk relatives. Genetic testing should be conducted in the context of pre- and post-test genetic counseling to ensure the patient's informed decision making. Patients who meet clinical criteria for a syndrome as well as those with identified pathogenic germline mutations should receive appropriate surveillance measures in order to minimize their overall risk of developing syndrome-specific cancers. This guideline specifically discusses genetic testing and management of Lynch syndrome, familial adenomatous polyposis (FAP), attenuated familial adenomatous polyposis (AFAP), MUTYH-associated polyposis (MAP), Peutz-Jeghers syndrome, juvenile polyposis syndrome, Cowden syndrome, serrated (hyperplastic) polyposis syndrome, hereditary pancreatic cancer, and hereditary gastric cancer.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Heritable somatic methylation and inactivation of MSH2 in families with Lynch syndrome due to deletion of the 3' exons of TACSTD1.

              Lynch syndrome patients are susceptible to colorectal and endometrial cancers owing to inactivating germline mutations in mismatch repair genes, including MSH2 (ref. 1). Here we describe patients from Dutch and Chinese families with MSH2-deficient tumors carrying heterozygous germline deletions of the last exons of TACSTD1, a gene directly upstream of MSH2 encoding Ep-CAM. Due to these deletions, transcription of TACSTD1 extends into MSH2. The MSH2 promoter in cis with the deletion is methylated in Ep-CAM positive but not in Ep-CAM negative normal tissues, thus revealing a correlation between activity of the mutated TACSTD1 allele and epigenetic inactivation of the corresponding MSH2 allele. Gene silencing by transcriptional read-through of a neighboring gene in either sense, as demonstrated here, or antisense direction, could represent a general mutational mechanism. Depending on the expression pattern of the neighboring gene that lacks its normal polyadenylation signal, this may cause either generalized or mosaic patterns of epigenetic inactivation.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Oncol
                Front Oncol
                Front. Oncol.
                Frontiers in Oncology
                Frontiers Media S.A.
                2234-943X
                24 May 2023
                2023
                : 13
                : 1141810
                Affiliations
                [1] 1 Department of Genetics, Grey Genetics , Brooklyn, NY, United States
                [2] 2 Research Department, Center for Genomic Interpretation , Sandy, UT, United States
                [3] 3 Cancer Genetics, Stanford Health Care , Palo Alto, CA, United States
                [4] 4 Center for Cancer Risk Assessment, Massachusetts General Hospital , Boston, MA, United States
                [5] 5 Cancer Prevention and Control Program, Fox Chase Cancer Center , Philadelphia, PA, United States
                [6] 6 Department of Gastroenterology, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona , Barcelona, Spain
                [7] 7 Division of Gastrointestinal Surgery, University of North Carolina-Chapel Hill , Chapel Hill, NC, United States
                [8] 8 Department of Internal Medicine, Huntsman Cancer Institute, University of Utah , Salt Lake City, UT, United States
                [9] 9 Department of Gastroenterology, MedStar Georgetown University Hospital , Washington, DC, United States
                [10] 10 Department of Gastroenterology, Kaiser Permanente Medical Center , Santa Clara, CA, United States
                [11] 11 Department of Colorectal Surgery and Sanford R. Weiss MD Center for Hereditary Colorectal Neoplasia, Digestive Disease and Surgery Institute, Cleveland Clinic , Cleveland, OH, United States
                [12] 12 Division of Gastroenterology, University of Arizona Cancer Center , Tucson, AZ, United States
                [13] 13 Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School , Boston, MA, United States
                [14] 14 Division of Medical Oncology, Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, CA, United States
                [15] 15 Department of Laboratory Medicine and Pathology, University of Washington , Seattle, WA, United States
                [16] 16 Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center , Houston, TX, United States
                [17] 17 Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA, United States
                [18] 18 Department of Tumor Biology, Institute of Cancer Research, The Norwegian Radium Hospital , Oslo, Norway
                Author notes

                Edited by: Gregory Idos, City of Hope National Medical Center, United States

                Reviewed by: Sergey Achkasov, Ryzhikh National Medical Research Centre of Coloproctology, Russia; Tao Huang, Chinese Academy of Sciences (CAS), China

                *Correspondence: Mev Dominguez-Valentin, mev.dominguez.valentin@ 123456rr-research.no
                Article
                10.3389/fonc.2023.1141810
                10247284
                37293588
                55ad5d0a-2c8d-4a54-9cc2-3dadf173cd4f
                Copyright © 2023 Mraz, Hodan, Rodgers-Fouche, Arora, Balaguer, Guillem, Jeter, Kanth, Li, Liska, Melson, Perez, Ricker, Shirts, Vilar, Katona and Dominguez-Valentin

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 10 January 2023
                : 02 May 2023
                Page count
                Figures: 6, Tables: 4, Equations: 0, References: 34, Pages: 15, Words: 5867
                Funding
                Funded by: Kreftforeningen , doi 10.13039/100008730;
                We acknowledge funding from the Norwegian Cancer Society, contract 194751-2017.
                Categories
                Oncology
                Original Research
                Custom metadata
                Gastrointestinal Cancers: Colorectal Cancer

                Oncology & Radiotherapy
                chemoprevention,crc,lynch syndrome,familial adenomatous polyposis,fap,barriers,aspirin

                Comments

                Comment on this article