1
|
Retsas S. Perceptions of Cancer Through the Ages-From Hippocratic Oncology to Precision Cancer Medicine. Biomolecules 2024; 14:1383. [PMID: 39595560 PMCID: PMC11591967 DOI: 10.3390/biom14111383] [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: 08/05/2024] [Revised: 09/13/2024] [Accepted: 10/14/2024] [Indexed: 11/28/2024] Open
Abstract
The unravelling of the human genome created new perceptions of the origin and evolution of diseases, and for cancer in particular, it established the notion that neoplasia has been a companion of life since its appearance on Earth. It is not surprising that neoplasms, in various forms, develop in numerous species of animals and even in plants. Unmistakable accounts of cancer with clinical features as are understood today begin in the 5th c. B.C. The principles and practice of the Hippocratic and Galenic tradition dominated cancer care virtually into the 20th century. Advanced sequencing technologies at the dawn of the 21st century generated new therapeutic opportunities with immunotherapy, oncolytic virotherapy, and gene transfer, with the latter especially being used in cases of hereditary cancer.
Collapse
Affiliation(s)
- Spyros Retsas
- Formerly Westminster Hospital, Chelsea and Westminster Hospital and Charing Cross Hospital, London SW10 9NH, UK
| |
Collapse
|
2
|
Li J, Yao H, Lu Y, Zhang S, Zhang Z. Chinese national clinical practice guidelines on prevention, diagnosis and treatment of early colorectal cancer. Chin Med J (Engl) 2024; 137:2017-2039. [PMID: 39104005 PMCID: PMC11374253 DOI: 10.1097/cm9.0000000000003253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND The incidence and mortality of colorectal cancer (CRC) in China are increasing in recent years. The clarified pathogenesis and detectable precancerous lesions of CRC make it possible to prevent, screen, and diagnose CRC at an early stage. With the development of endoscopic and surgical techniques, the choice of treatment for early CRC is also worth further discussion, and accordingly, a standard follow-up program after treatment needs to be established. METHODS This clinical practice guideline (CPG) was developed following the recommended process of the World Health Organization, adopting Grading of Recommendations Assessment, Development and Evaluation (GRADE) in assessing evidence quality, and using the Evidence to Decision framework to formulate clinical recommendations, thereby minimizing bias and increasing transparency of the CPG development process. We used the Reporting Items for practice Guidelines in HealThcare (RIGHT) statement and Appraisal of Guidelines for Research and Evaluation II (AGREE II) as reporting and conduct guides to ensure the guideline's completeness and transparency. RESULTS This CPG comprises 46 recommendations concerning prevention, screening, diagnosis, treatment, and surveillance of CRC. In these recommendations, we have indicated protective and risk factors for CRC and made recommendations for chemoprevention. We proposed a suitable screening program for CRC based on the Chinese context. We also provided normative statements for the diagnosis, treatment, and surveillance of CRC based on existing clinical evidence and guidelines. CONCLUSIONS The 46 recommendations in this CPG are formed with consideration for stakeholders' values and preferences, feasibility, and acceptability. Recommendations are generalizable to resource-limited settings with similar CRC epidemiology pattern as China.
Collapse
Affiliation(s)
- Jingnan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Hongwei Yao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Diseases, Beijing 100050, China
| | - Yun Lu
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, China
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing 100050, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Diseases, Beijing 100050, China
| |
Collapse
|
3
|
Fu QQ, Ma L, Niu XM, Zhao HX, Ge XH, Jin H, Yu DH, Yang S. Trends and hotspots in gastrointestinal neoplasms risk assessment: A bibliometric analysis from 1984 to 2022. World J Gastrointest Oncol 2024; 16:2842-2861. [PMID: 38994129 PMCID: PMC11236220 DOI: 10.4251/wjgo.v16.i6.2842] [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: 01/12/2024] [Revised: 03/10/2024] [Accepted: 04/15/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Gastrointestinal neoplasm (GN) significantly impact the global cancer burden and mortality, necessitating early detection and treatment. Understanding the evolution and current state of research in this field is vital. AIM To conducts a comprehensive bibliometric analysis of publications from 1984 to 2022 to elucidate the trends and hotspots in the GN risk assessment research, focusing on key contributors, institutions, and thematic evolution. METHODS This study conducted a bibliometric analysis of data from the Web of Science Core Collection database using the "bibliometrix" R package, VOSviewer, and CiteSpace. The analysis focused on the distribution of publications, contributions by institutions and countries, and trends in keywords. The methods included data synthesis, network analysis, and visualization of international collaboration networks. RESULTS This analysis of 1371 articles on GN risk assessment revealed a notable evolution in terms of research focus and collaboration. It highlights the United States' critical role in advancing this field, with significant contributions from institutions such as Brigham and Women's Hospital and the National Cancer Institute. The last five years, substantial advancements have been made, representing nearly 45% of the examined literature. Publication rates have dramatically increased, from 20 articles in 2002 to 112 in 2022, reflecting intensified research efforts. This study underscores a growing trend toward interdisciplinary and international collaboration, with the Journal of Clinical Oncology standing out as a key publication outlet. This shift toward more comprehensive and collaborative research methods marks a significant step in addressing GN risks. CONCLUSION This study underscores advancements in GN risk assessment through genetic analyses and machine learning and reveals significant geographical disparities in research emphasis. This calls for enhanced global collaboration and integration of artificial intelligence to improve cancer prevention and treatment accuracy, ultimately enhancing worldwide patient care.
Collapse
Affiliation(s)
- Qiang-Qiang Fu
- Department of General Practice, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200090, China
- Shanghai General Practice and Community Health Development Research Center, Shanghai 200090, China
| | - Le Ma
- Department of General Practice, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200090, China
| | - Xiao-Min Niu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Hua-Xin Zhao
- Department of Oncology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200090, China
| | - Xu-Hua Ge
- Department of General Practice, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200090, China
| | - Hua Jin
- Department of General Practice, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200090, China
| | - De-Hua Yu
- Department of General Practice, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200090, China
- Shanghai General Practice and Community Health Development Research Center, Shanghai 200090, China
| | - Sen Yang
- Department of General Practice, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200090, China
- Shanghai General Practice and Community Health Development Research Center, Shanghai 200090, China
| |
Collapse
|
4
|
Lee NY, Hum M, Wong M, Ong PY, Lee SC, Lee ASG. Alleviating misclassified germline variants in underrepresented populations: A strategy using popmax. Genet Med 2024; 26:101124. [PMID: 38522067 DOI: 10.1016/j.gim.2024.101124] [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: 09/08/2023] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024] Open
Abstract
PURPOSE Germline variant interpretation often depends on population-matched control cohorts. This is not feasible for population groups that are underrepresented in current population reference databases. METHODS We classify germline variants with population-matched controls for 2 ancestrally diverse cohorts of patients: 132 early-onset or familial colorectal carcinoma patients from Singapore and 100 early-onset colorectal carcinoma patients from the United States. The effects of using a population-mismatched control cohort are simulated by swapping the control cohorts used for each patient cohort, with or without the popmax computational strategy. RESULTS Population-matched classifications revealed a combined 62 pathogenic or likely pathogenic (P/LP) variants in 34 genes across both cohorts. Using a population-mismatched control cohort resulted in misclassification of non-P/LP variants as P/LP, driven by the absence of ancestry-specific rare variants in the control cohort. Popmax was more effective in alleviating misclassifications for the Singapore cohort than the US cohort. CONCLUSION Underrepresented population groups can suffer from higher rates of false-positive P/LP results. Popmax can partially alleviate these misclassifications, but its efficacy still depends on the degree with which the population groups are represented in the control cohort.
Collapse
Affiliation(s)
- Ning Yuan Lee
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore
| | - Melissa Hum
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore
| | - Matthew Wong
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore
| | - Pei-Yi Ong
- Department of Hematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore
| | - Soo-Chin Lee
- Department of Hematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cancer Science Institute, Singapore (CSI), National University of Singapore, Singapore
| | - Ann S G Lee
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore; SingHealth Duke-NUS Oncology Academic Clinical Programme (ONCO ACP), Duke-NUS Medical School, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
5
|
Shao B, Di L. Metastatic breast cancer with double heterozygosity for the BRCA1 and BRCA2 genes responding to olaparib: A case report. Oncol Lett 2024; 27:253. [PMID: 38646498 PMCID: PMC11027096 DOI: 10.3892/ol.2024.14387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/07/2023] [Indexed: 04/23/2024] Open
Abstract
Olaparib was the first poly ADP-ribose polymerase inhibitor approved for patients with cancer with mutations in either BRCA1 or BRCA2 in China. To the best of our knowledge, however, no study has described the efficacy of olaparib for patients with breast cancer with double mutations in BRCA1 and BRCA2. The present case report describes a patient with breast cancer with deleterious germline mutations in both BRCA1 and BRCA2. The 56-year-old patient with multiple metastatic breast cancer underwent breast cancer resection with 12 years interval between removal of the left and right breast. Germline mutations in both BRCA1 (S405X) and BRCA2 (W2990X) were identified by NGS. She received two cycles of chemotherapy with a combination of albumin-bound paclitaxel and capecitabine; the response was progressive disease. Subsequently, the patient was treated with a gradual dosage of decreasing olaparib (600 to 300 mg BID) for 6 months until grade 3 anemia could not be alleviated by giving erythropoietin and iron, and CT imaging showed a partial response (35% reduction). The patient then switched to exemestane therapy due to the continuous grade 3 anemia. In conclusion, the present study reported a female patient with double heterozygosity of BRCA1 and BRCA2 who benefited from olaparib monotherapy. Thus, olaparib may be a suitable treatment for such patients.
Collapse
Affiliation(s)
- Bin Shao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Lijun Di
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| |
Collapse
|
6
|
Zohud O, Midlej K, Lone IM, Nashef A, Abu-Elnaaj I, Iraqi FA. Studying the Effect of the Host Genetic Background of Juvenile Polyposis Development Using Collaborative Cross and Smad4 Knock-Out Mouse Models. Int J Mol Sci 2024; 25:5812. [PMID: 38891999 PMCID: PMC11172477 DOI: 10.3390/ijms25115812] [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: 03/28/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Juvenile polyposis syndrome (JPS) is a rare autosomal dominant disorder characterized by multiple juvenile polyps in the gastrointestinal tract, often associated with mutations in genes such as Smad4 and BMPR1A. This study explores the impact of Smad4 knock-out on the development of intestinal polyps using collaborative cross (CC) mice, a genetically diverse model. Our results reveal a significant increase in intestinal polyps in Smad4 knock-out mice across the entire population, emphasizing the broad influence of Smad4 on polyposis. Sex-specific analyses demonstrate higher polyp counts in knock-out males and females compared to their WT counterparts, with distinct correlation patterns. Line-specific effects highlight the nuanced response to Smad4 knock-out, underscoring the importance of genetic variability. Multimorbidity heat maps offer insights into complex relationships between polyp counts, locations, and sizes. Heritability analysis reveals a significant genetic basis for polyp counts and sizes, while machine learning models, including k-nearest neighbors and linear regression, identify key predictors, enhancing our understanding of juvenile polyposis genetics. Overall, this study provides new information on understanding the intricate genetic interplay in the context of Smad4 knock-out, offering valuable insights that could inform the identification of potential therapeutic targets for juvenile polyposis and related diseases.
Collapse
Affiliation(s)
- Osayd Zohud
- Department of Clinical Microbiology and Immunology, Faculty of Medicine and Health Sciences, Tel-Aviv University, Tel-Aviv 69978, Israel; (O.Z.); (K.M.); (I.M.L.)
| | - Kareem Midlej
- Department of Clinical Microbiology and Immunology, Faculty of Medicine and Health Sciences, Tel-Aviv University, Tel-Aviv 69978, Israel; (O.Z.); (K.M.); (I.M.L.)
| | - Iqbal M. Lone
- Department of Clinical Microbiology and Immunology, Faculty of Medicine and Health Sciences, Tel-Aviv University, Tel-Aviv 69978, Israel; (O.Z.); (K.M.); (I.M.L.)
| | - Aysar Nashef
- Department of Oral and Maxillofacial Surgery, Baruch Padeh Medical Center, Poriya Tebaria 42310, Israel; (A.N.); (I.A.-E.)
- Department of Oral and Maxillofacial Surgery, Meir Medical Center, Faculty of Medicine and Health Sciences, Tel-Aviv University, Kfar-Saba 69978, Israel
| | - Imad Abu-Elnaaj
- Department of Oral and Maxillofacial Surgery, Baruch Padeh Medical Center, Poriya Tebaria 42310, Israel; (A.N.); (I.A.-E.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Tsaft 33241, Israel
| | - Fuad A. Iraqi
- Department of Clinical Microbiology and Immunology, Faculty of Medicine and Health Sciences, Tel-Aviv University, Tel-Aviv 69978, Israel; (O.Z.); (K.M.); (I.M.L.)
| |
Collapse
|
7
|
Horton C, Hoang L, Zimmermann H, Young C, Grzybowski J, Durda K, Vuong H, Burks D, Cass A, LaDuca H, Richardson ME, Harrison S, Chao EC, Karam R. Diagnostic Outcomes of Concurrent DNA and RNA Sequencing in Individuals Undergoing Hereditary Cancer Testing. JAMA Oncol 2024; 10:212-219. [PMID: 37924330 PMCID: PMC10625669 DOI: 10.1001/jamaoncol.2023.5586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/04/2023] [Indexed: 11/06/2023]
Abstract
Importance Personalized surveillance, prophylaxis, and cancer treatment options for individuals with hereditary cancer predisposition are informed by results of germline genetic testing. Improvements to genomic technology, such as the availability of RNA sequencing, may increase identification of individuals eligible for personalized interventions by improving the accuracy and yield of germline testing. Objective To assess the cumulative association of paired DNA and RNA testing with detection of disease-causing germline genetic variants and resolution of variants of uncertain significance (VUS). Design, Setting, and Participants Paired DNA and RNA sequencing was performed on individuals undergoing germline testing for hereditary cancer indication at a single diagnostic laboratory from March 2019 through April 2020. Demographic characteristics, clinical data, and test results were curated as samples were received, and changes to variant classification were assessed over time. Data analysis was performed from May 2020 to June 2023. Main Outcomes and Measures Main outcomes were increase in diagnostic yield, decrease in VUS rate, the overall results by variant type, the association of RNA evidence with variant classification, and the corresponding predicted effect on cancer risk management. Results A total of 43 524 individuals were included (median [range] age at testing, 54 [2-101] years; 37 373 female individuals [85.7%], 6224 male individuals [14.3%], and 2 individuals of unknown sex [<0.1%]), with 43 599 tests. A total of 2197 (5.0%) were Ashkenazi Jewish, 1539 (3.5%) were Asian, 3077 (7.1%) were Black, 2437 (5.6%) were Hispanic, 27 793 (63.7%) were White, and 2049 (4.7%) were other race, and for 4507 individuals (10.3%), race and ethnicity were unknown. Variant classification was impacted in 549 individuals (1.3%). Medically significant upgrades were made in 97 individuals, including 70 individuals who had a variant reclassified from VUS to pathogenic/likely pathogenic (P/LP) and 27 individuals who had a novel deep intronic P/LP variant that would not have been detected using DNA sequencing alone. A total of 93 of 545 P/LP splicing variants (17.1%) were dependent on RNA evidence for classification, and 312 of 439 existing splicing VUS (71.1%) were resolved by RNA evidence. Notably, the increase in positive rate (3.1%) and decrease in VUS rate (-3.9%) was higher in Asian, Black, and Hispanic individuals combined compared to White individuals (1.6%; P = .02; and -2.5%; P < .001). Conclusions and Relevance Findings of this diagnostic study demonstrate that the ability to perform RNA sequencing concurrently with DNA sequencing represents an important advancement in germline genetic testing by improving detection of novel variants and classification of existing variants. This expands the identification of individuals with hereditary cancer predisposition and increases opportunities for personalization of therapeutics and surveillance.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huy Vuong
- Ambry Genetics, Aliso Viejo, California
| | | | | | | | | | | | - Elizabeth C. Chao
- Ambry Genetics, Aliso Viejo, California
- University of California, Irvine, School of Medicine
| | | |
Collapse
|
8
|
Iwamuro M, Kawano S, Otsuka M. Differential Diagnoses and Management Approaches for Gastric Polyposis. GASTROENTEROLOGY INSIGHTS 2024; 15:122-144. [DOI: 10.3390/gastroent15010009] [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: 01/06/2025] Open
Abstract
Multiple gastric polyps are observed in various polyposis syndromes and conditions associated with polypoid lesion development in the stomach. Polyposis syndromes often occur concurrently with specific malignant tumors and can manifest at any point in an individual’s lifespan, thus explaining the diversity in surveillance methods. Furthermore, genetic counseling and surveillance are essential not only for the patients themselves but also for their blood relatives. Therefore, the accurate diagnosis and appropriate surveillance of multiple gastric polyps are crucial for improving patient outcomes. This review aims to provide essential information on such lesions along with representative endoscopic images of familial adenomatous polyposis, Peutz-Jeghers syndrome, Cowden syndrome, Cronkhite-Canada syndrome, juvenile polyposis syndrome, gastric adenocarcinoma and proximal polyposis of the stomach, neuroendocrine tumors in autoimmune gastritis, proton pump inhibitor-related gastric mucosal changes, and multiple submucosal heterotopic glands. We wish for this review to serve as a valuable resource for endoscopists seeking to deepen their comprehension of gastric polyposis.
Collapse
Affiliation(s)
- Masaya Iwamuro
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-0082, Japan
| | - Seiji Kawano
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-0082, Japan
| | - Motoyuki Otsuka
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-0082, Japan
| |
Collapse
|
9
|
Brand RM, Dudley B, Karloski E, Zyhowski A, Raphael R, Pitlor D, Metter EJ, Pai R, Lee K, Brand RE, Uttam S. Immune microenvironment profiling of normal appearing colorectal mucosa biopsied over repeat patient visits reproducibly separates lynch syndrome patients based on their history of colon cancer. Front Oncol 2023; 13:1174831. [PMID: 37637062 PMCID: PMC10457127 DOI: 10.3389/fonc.2023.1174831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 07/14/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction Lynch syndrome (LS) is the most common hereditary cause of colorectal cancer (CRC), increasing lifetime risk of CRC by up to 70%. Despite this higher lifetime risk, disease penetrance in LS patients is highly variable and most LS patients undergoing CRC surveillance will not develop CRC. Therefore, biomarkers that can correctly and consistently predict CRC risk in LS patients are needed to both optimize LS patient surveillance and help identify better prevention strategies that reduce risk of CRC development in the subset of high-risk LS patients. Methods Normal-appearing colorectal tissue biopsies were obtained during repeat surveillance colonoscopies of LS patients with and without a history of CRC, healthy controls (HC), and patients with a history of sporadic CRC. Biopsies were cultured in an ex-vivo explant system and their supernatants were assayed via multiplexed ELISA to profile the local immune signaling microenvironment. High quality cytokines were identified using the rxCOV fidelity metric. These cytokines were used to perform elastic-net penalized logistic regression-based biomarker selection by computing a new measure - overall selection probability - that quantifies the ability of each marker to discriminate between patient cohorts being compared. Results Our study demonstrated that cytokine based local immune microenvironment profiling was reproducible over repeat visits and sensitive to patient LS-status and CRC history. Furthermore, we identified sets of cytokines whose differential expression was predictive of LS-status in patients when compared to sporadic CRC patients and in identifying those LS patients with or without a history of CRC. Enrichment analysis based on these biomarkers revealed an LS and CRC status dependent constitutive inflammatory state of the normal appearing colonic mucosa. Discussion This prospective pilot study demonstrated that immune profiling of normal appearing colonic mucosa discriminates LS patients with a prior history of CRC from those without it, as well as patients with a history of sporadic CRC from HC. Importantly, it suggests the existence of immune signatures specific to LS-status and CRC history. We anticipate that our findings have the potential to assess CRC risk in individuals with LS and help in preemptively mitigating it by optimizing surveillance and identifying candidate prevention targets. Further studies are required to validate our findings in an independent cohort of LS patients over multiple visits.
Collapse
Affiliation(s)
- Rhonda M. Brand
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Magee Womens Research Institute, Pittsburgh, PA, United States
| | - Beth Dudley
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Eve Karloski
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ashley Zyhowski
- Magee Womens Research Institute, Pittsburgh, PA, United States
| | - Rebecca Raphael
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, United States
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Danielle Pitlor
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - E. Jeffrey Metter
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Reet Pai
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kenneth Lee
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Randall E. Brand
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Shikhar Uttam
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, United States
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
10
|
Zhang L, Zheng Y, Yu X, Yu K, Zhu S. Primary intra‑abdominal desmoid fibromatosis associated with familial adenomatous polyposis: A case report. Exp Ther Med 2023; 26:308. [PMID: 37273755 PMCID: PMC10236137 DOI: 10.3892/etm.2023.12007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 02/15/2023] [Indexed: 06/06/2023] Open
Abstract
Desmoid fibromatosis (DF) is a clonal proliferative disorder of myofibroblasts, which arises, with a low incidence, in soft tissue, including within the abdomen. The incidence of DF is associated with familial adenomatous polyposis (FAP), and is more common following FAP surgery. It is rare for a patient to make his/her first visit to hospital due to DF symptoms associated with FAP. In the present report, a case of mesenteric DF associated with FAP is described. This case also had incomplete intestinal obstruction due to DF. By summarizing previous studies examining DF and FAP treatment, combined with the disease characteristics of this patient, the clinical treatment strategy for DF associated with FAP was explored.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Hepatobiliary Surgery, Weihai Central Hospital, Weihai, Shandong 264200, P.R. China
| | - Yaotun Zheng
- Department of Anesthesia, Weihai Central Hospital, Weihai, Shandong 264200, P.R. China
| | - Xiaoqian Yu
- Department of Obstetrics, Weihai Central Hospital, Weihai, Shandong 264200, P.R. China
| | - Kang Yu
- Department of Research and Development, Tianjin Lianchuang Technology Development Co., Ltd., Tianjin 300000, P.R. China
| | - Shengjie Zhu
- Department of Oncology, Weihai Central Hospital, Weihai, Shandong 264200, P.R. China
| |
Collapse
|
11
|
Aberrant HMGA2 Expression Sustains Genome Instability That Promotes Metastasis and Therapeutic Resistance in Colorectal Cancer. Cancers (Basel) 2023; 15:cancers15061735. [PMID: 36980621 PMCID: PMC10046046 DOI: 10.3390/cancers15061735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/06/2023] [Accepted: 03/11/2023] [Indexed: 03/16/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most lethal cancers worldwide, accounting for nearly ~10% of all cancer diagnoses and deaths. Current therapeutic approaches have considerably increased survival for patients diagnosed at early stages; however, ~20% of CRC patients are diagnosed with late-stage, metastatic CRC, where 5-year survival rates drop to 6–13% and treatment options are limited. Genome instability is an enabling hallmark of cancer that confers increased acquisition of genetic alterations, mutations, copy number variations and chromosomal rearrangements. In that regard, research has shown a clear association between genome instability and CRC, as the accumulation of aberrations in cancer-related genes provides subpopulations of cells with several advantages, such as increased proliferation rates, metastatic potential and therapeutic resistance. Although numerous genes have been associated with CRC, few have been validated as predictive biomarkers of metastasis or therapeutic resistance. A growing body of evidence suggests a member of the High-Mobility Group A (HMGA) gene family, HMGA2, is a potential biomarker of metastatic spread and therapeutic resistance. HMGA2 is expressed in embryonic tissues and is frequently upregulated in aggressively growing cancers, including CRC. As an architectural, non-histone chromatin binding factor, it initiates chromatin decompaction to facilitate transcriptional regulation. HMGA2 maintains the capacity for stem cell renewal in embryonic and cancer tissues and is a known promoter of epithelial-to-mesenchymal transition in tumor cells. This review will focus on the known molecular mechanisms by which HMGA2 exerts genome protective functions that contribute to cancer cell survival and chemoresistance in CRC.
Collapse
|
12
|
Brand RM, Dudley B, Karloski E, Zyhowski A, Raphael R, Pitlor D, Metter EJ, Pai R, Lee K, Brand RE, Uttam S. Immune Microenvironment Profiling of Normal Appearing Colorectal Mucosa Biopsied Over Repeat Patient Visits Reproduciably Separates Lynch Syndrome Patients Based on Their History of Colon Cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.03.23286594. [PMID: 36945451 PMCID: PMC10029019 DOI: 10.1101/2023.03.03.23286594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
Introduction Lynch syndrome (LS) is the most common hereditary cause of colorectal cancer (CRC), increasing lifetime risk of CRC by up to 70%. Despite this higher lifetime risk, disease penetrance in LS patients is highly variable and most LS patients undergoing CRC surveillance will not develop CRC. Therefore, biomarkers that can correctly and consistently predict CRC risk in LS patients are needed to both optimize LS patient surveillance and help identify better prevention strategies that reduce risk of CRC development in the subset of high-risk LS patients. Methods Normal-appearing colorectal tissue biopsies were obtained during repeat surveillance colonoscopies of LS patients with and without a history of CRC, healthy controls (HC), and patients with a history of sporadic CRC. Biopsies were cultured in an ex-vivo explant system and their supernatants were assayed via multiplexed ELISA to profile the local immune signaling microenvironment. High quality cytokine signatures were identified using rx COV fidelity metric. These signatures were used to perform biomarker selection by computing their selection probability based on penalized logistic regression. Results Our study demonstrated that cytokine based local immune microenvironment profiling was reproducible over repeat visits and sensitive to patient LS-status and CRC history. Furthermore, we identified sets of biomarkers whose differential expression was predictive of LS-status in patients when compared to sporadic CRC patients and in identifying those LS patients with or without a history of CRC. Enrichment analysis based on these biomarkers revealed an LS and CRC status dependent constitutive inflammatory state of the normal appearing colonic mucosa. Discussion This prospective pilot study demonstrated that immune profiling of normal appearing colonic mucosa discriminates LS patients with a prior history of CRC from those without it, as well as patients with a history of sporadic CRC from HC. Importantly, it suggests existence of immune signatures specific to LS-status and CRC history. We anticipate that our findings have the potential to assess CRC risk in individuals with LS and help in preemptively mitigating it by optimizing surveillance and identifying candidate prevention targets. Further studies are required to validate our findings in an independent cohort of LS patients over multiple visits.
Collapse
|
13
|
Imyanitov EN, Kuligina ES, Sokolenko AP, Suspitsin EN, Yanus GA, Iyevleva AG, Ivantsov AO, Aleksakhina SN. Hereditary cancer syndromes. World J Clin Oncol 2023; 14:40-68. [PMID: 36908677 PMCID: PMC9993141 DOI: 10.5306/wjco.v14.i2.40] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/09/2022] [Accepted: 02/14/2023] [Indexed: 02/21/2023] Open
Abstract
Hereditary cancer syndromes (HCSs) are arguably the most frequent category of Mendelian genetic diseases, as at least 2% of presumably healthy subjects carry highly-penetrant tumor-predisposing pathogenic variants (PVs). Hereditary breast-ovarian cancer and Lynch syndrome make the highest contribution to cancer morbidity; in addition, there are several dozen less frequent types of familial tumors. The development of the majority albeit not all hereditary malignancies involves two-hit mechanism, i.e. the somatic inactivation of the remaining copy of the affected gene. Earlier studies on cancer families suggested nearly fatal penetrance for the majority of HCS genes; however, population-based investigations and especially large-scale next-generation sequencing data sets demonstrate that the presence of some highly-penetrant PVs is often compatible with healthy status. Hereditary cancer research initially focused mainly on cancer detection and prevention. Recent studies identified multiple HCS-specific drug vulnerabilities, which translated into the development of highly efficient therapeutic options.
Collapse
Affiliation(s)
- Evgeny N Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Ekaterina S Kuligina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Anna P Sokolenko
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Evgeny N Suspitsin
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Grigoriy A Yanus
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Aglaya G Iyevleva
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Alexandr O Ivantsov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Svetlana N Aleksakhina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| |
Collapse
|
14
|
Vaccaro CA, Soarez J, Dominguez-Valentin M, Pavicic WH. Editorial: Advances in genetics and molecular diagnosis in colorectal, stomach, and pancreatic cancer. Front Oncol 2022; 12:1104671. [PMID: 36601476 PMCID: PMC9806422 DOI: 10.3389/fonc.2022.1104671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Affiliation(s)
- Carlos Alberto Vaccaro
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB) Hospital Italiano de Buenos Aires, Instituto Universitario Hospital Italiano de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (HIBA-IUHI-CONICET), Ciudad Autónoma de Buenos Aires (CABA), Argentina,Programa de Cáncer Hereditario (Pro.Can.He.), Hospital Italiano de Buenos Aires (HIBA), Ciudad Autónoma de Buenos Aires (CABA), Argentina
| | - Julieta Soarez
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB) Hospital Italiano de Buenos Aires, Instituto Universitario Hospital Italiano de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (HIBA-IUHI-CONICET), Ciudad Autónoma de Buenos Aires (CABA), Argentina,Programa de Cáncer Hereditario (Pro.Can.He.), Hospital Italiano de Buenos Aires (HIBA), Ciudad Autónoma de Buenos Aires (CABA), Argentina
| | - Mev Dominguez-Valentin
- Department of Tumor Biology, Institute of Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
| | - Walter Hernán Pavicic
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB) Hospital Italiano de Buenos Aires, Instituto Universitario Hospital Italiano de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (HIBA-IUHI-CONICET), Ciudad Autónoma de Buenos Aires (CABA), Argentina,Programa de Cáncer Hereditario (Pro.Can.He.), Hospital Italiano de Buenos Aires (HIBA), Ciudad Autónoma de Buenos Aires (CABA), Argentina,*Correspondence: Walter Hernán Pavicic,
| |
Collapse
|
15
|
Chen L, Ye L, Hu B. Hereditary Colorectal Cancer Syndromes: Molecular Genetics and Precision Medicine. Biomedicines 2022; 10:biomedicines10123207. [PMID: 36551963 PMCID: PMC9776295 DOI: 10.3390/biomedicines10123207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/28/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) is the third most commonly diagnosed cancer worldwide. Hereditary CRC syndromes account for approximately 5-10% of all CRC, with a lifetime risk of CRC that approaches 50-80% in the absence of endoscopic or surgical treatment. Hereditary CRC syndromes can be phenotypically divided into polyposis and non-polyposis syndrome, mainly according to the conditions of polyps. The typical representatives are familial adenomatous polyposis (FAP) and Lynch syndromes (LS), respectively. Over the past few decades, molecular genetics enhanced the discovery of cancer-predisposing genes and revolutionized the field of clinical oncology. Hereditary CRC syndromes have been a key part of this effort, with data showing that pathogenic variants are present in up to 10% of cases. Molecular phenotypes of tumors can not only help identify individuals with genetic susceptibility to CRC but also guide the precision prevention and treatment for the development of CRC. This review emphasizes the molecular basis and prevention strategies for hereditary CRC syndromes.
Collapse
Affiliation(s)
| | | | - Bing Hu
- Correspondence: ; Tel.: +86-18980601278
| |
Collapse
|
16
|
Kastner AM, Kocak HK, Fischer-Jacobs J, Hahne A, Zimmermann T. Desire for Children and Distress in Women with Hereditary Cancer Syndromes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:14517. [PMID: 36361396 PMCID: PMC9654350 DOI: 10.3390/ijerph192114517] [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: 09/24/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 06/16/2023]
Abstract
The diagnosis of a hereditary cancer syndrome can be psychologically stressful and influence family planning. This study aimed to gain insights into the relationship between the desire for children and the distress of female carriers. Women (N = 255) with different hereditary cancer syndromes were assessed from November 2019 to July 2021 at genetic counseling centers, the centers of the German HBOC-Consortium and the centers of the German HNPCC-Consortium regarding their distress levels with the NCCN Distress Thermometer (DT). The desire for children was measured by self-developed questions. Levels of distress and desire for children were evaluated descriptively. Factors influencing the desire for children and distress were calculated using binary logistic regression: 56% (n = 51) of 18- to 39-year-old participants reported a desire to have children; 70.6% of the carriers with a desire for children indicated a need for advice from their physicians regarding family planning. The diagnosis led 61.5% to postpone the timing of family planning, and the majority (68.8%) opted for an earlier birth. Carriers had higher levels of distress. Younger carriers (p = 0.037) and those living in poorer economic circumstances (p = 0.011) were more distressed. The diagnosis of hereditary cancer syndrome affects family planning. The results emphasize the importance of physicians addressing family planning in their counseling sessions.
Collapse
Affiliation(s)
- Anna Maria Kastner
- Department of Psychosomatic Medicine and Psychotherapy, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Hatice Kübra Kocak
- Department of Psychosomatic Medicine and Psychotherapy, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Josefine Fischer-Jacobs
- Department of Psychosomatic Medicine and Psychotherapy, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Andrea Hahne
- BRCA-Netzwerk e.V., Thomas-Mann-Street 40, 53111 Bonn, Germany
| | - Tanja Zimmermann
- Department of Psychosomatic Medicine and Psychotherapy, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| |
Collapse
|
17
|
A Previously Unrecognized Molecular Landscape of Lynch Syndrome in the Mexican Population. Int J Mol Sci 2022; 23:ijms231911549. [PMID: 36232851 PMCID: PMC9569652 DOI: 10.3390/ijms231911549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/26/2022] [Accepted: 09/09/2022] [Indexed: 11/30/2022] Open
Abstract
Lynch syndrome (LS) is the main hereditary colorectal cancer syndrome. There have been few reports regarding the clinical and molecular characteristics of LS patients in Latin America; this is particularly true in the Mexican population, where no information is available. The present study aims to describe the clinical and molecular spectrum of variants in a cohort of patients diagnosed with LS in Mexico. We present a retrospective analysis of 412 patients with suspected LS, whose main site of cancer diagnosis was the colon (58.25%), followed by the endometrium (18.93%). Next-generation sequencing analysis, with an extensive multigene panel, showed that 27.1% (112/414) had a variant in one of the genes of the mismatch repair pathway (MMR); 30.4% (126/414) had a variant in non-MMR genes such as CHEK2, APC, MUTYH, BRCA1, and BRCA2; and 42.5% (176/414) had no genetic variants. Most of the variants were found in MLH1. Pathogenic variants (PVs) in MMR genes were identified in 65.7% (96/146) of the total PVs, and 34.24% (45/146) were in non-MMR genes. Molecular and clinical characterization of patients with LS in specific populations allowed personalized follow-up, with the option for targeted treatment with immune checkpoint inhibitors and the development of public health policies. Moreover, such characterization allows for family cascade testing and consequent prevention strategies.
Collapse
|
18
|
Chang Y, Chao D, Chung C, Chou Y, Chang C, Lin C, Chu H, Chen H, Liu T, Juan Y, Chang S, Chang J. Cancer carrier screening in the general population using whole-genome sequencing. Cancer Med 2022; 12:1972-1983. [PMID: 35861108 PMCID: PMC9883534 DOI: 10.1002/cam4.5034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Cancer is a major cause of death, and its early identification and intervention have potential for clinical actionability and benefits for human health. The studies using whole-genome sequencing (WGS) and large samples analysis of cancer-related genes have been rarely done. METHODS We performed WGS to explore germline mutations in coding and non-coding areas of cancer-related genes and non-coding driver genes and regulatory areas. Structural variants (SVs) was also analyzed. We used several tools and a subgrouping method to analyze the variants in 1491 healthy participants. Moreover, 275 cancer-related genes sequencing was carried out in 125 cancer patients. RESULTS The incidence of familial cancer in the Taiwanese general population is 8.79% (131/1491). Cancer carrier rate of cancer-related genes is about 7.04% (105/1491) for pathogenic/likely pathogenic variants (P/LP) on ClinVar database only, and 28.24% (421/1491) for P/LP and loss of function variants. The carrier frequencies of cancer-related genes P/LP on ClinVar database were as follows: 8.40% (11/131), 7.11% (28/394), and 6.83% (66/966) in FC, 1MC, and nMC, respectively. The SVs and non-coding driver gene variants are uncommon. There are 1.54% (23/1491) of actionable cancer genes in American College of Medical Genetics and Genomics (ACMG), and the germline mutation rate of 275 cancer-related genes is 7.2% (9/125) in cancer patients including 4.0% (5/125) of actionable cancer genes in ACMG. After analyzing the frequencies of P/LP variants on GJB2 and SLC25A13 genes, we suggest that these two genes may not be cancer-related genes and need be re-evaluated. CONCLUSIONS WGS analysis can completely detect germline mutations in cancer carriers. This study use subgrouping approach for samples provides a strategy to study whether a gene or variant is a cancer-related gene or variant in the future studies.
Collapse
Affiliation(s)
- Ya‐Sian Chang
- Center for Precision MedicineChina Medical University HospitalTaichungTaiwan
- Epigenome Research CenterChina Medical University HospitalTaichungTaiwan
- Department of Laboratory MedicineChina Medical University HospitalTaichungTaiwan
- School of MedicineChina Medical UniversityTaichungTaiwan
| | - Dy‐San Chao
- Center for Precision MedicineChina Medical University HospitalTaichungTaiwan
- Epigenome Research CenterChina Medical University HospitalTaichungTaiwan
- Department of Laboratory MedicineChina Medical University HospitalTaichungTaiwan
| | - Chin‐Chun Chung
- Center for Precision MedicineChina Medical University HospitalTaichungTaiwan
| | - Yu‐Pao Chou
- Center for Precision MedicineChina Medical University HospitalTaichungTaiwan
- Epigenome Research CenterChina Medical University HospitalTaichungTaiwan
- Department of Laboratory MedicineChina Medical University HospitalTaichungTaiwan
| | - Chieh‐Min Chang
- Center for Precision MedicineChina Medical University HospitalTaichungTaiwan
- Epigenome Research CenterChina Medical University HospitalTaichungTaiwan
- Department of Laboratory MedicineChina Medical University HospitalTaichungTaiwan
| | - Chia‐Li Lin
- Center for Precision MedicineChina Medical University HospitalTaichungTaiwan
| | - Hou‐Wei Chu
- Institute of Biomedical Sciences|Academia SinicaTaipeiTaiwan
| | - Hon‐Da Chen
- Center for Precision MedicineChina Medical University HospitalTaichungTaiwan
- Epigenome Research CenterChina Medical University HospitalTaichungTaiwan
- Department of Laboratory MedicineChina Medical University HospitalTaichungTaiwan
| | - Ting‐Yuan Liu
- Center for Precision MedicineChina Medical University HospitalTaichungTaiwan
| | - Yu‐Hsuan Juan
- Center for Precision MedicineChina Medical University HospitalTaichungTaiwan
| | - Shun‐Jen Chang
- Department of Kinesiology, Health and Leisure StudiesNational University of KaohsiungKaohsiungTaiwan
| | - Jan‐Gowth Chang
- Center for Precision MedicineChina Medical University HospitalTaichungTaiwan
- Epigenome Research CenterChina Medical University HospitalTaichungTaiwan
- Department of Laboratory MedicineChina Medical University HospitalTaichungTaiwan
- School of MedicineChina Medical UniversityTaichungTaiwan
- Department of Bioinformatics and Medical EngineeringAsia UniversityTaichungTaiwan
| |
Collapse
|
19
|
Mansur A, Zhang F, Lu CY. Genetic Testing and/or Counseling for Colorectal Cancer by Health Insurance Type. J Pers Med 2022; 12:jpm12071146. [PMID: 35887643 PMCID: PMC9317363 DOI: 10.3390/jpm12071146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/03/2022] [Accepted: 07/13/2022] [Indexed: 12/18/2022] Open
Abstract
Genetic testing is increasingly used in clinical practice to assist with the diagnosis of genetic diseases and/or provide information about disease risk, and genetic counseling supports patient understanding of test results before and/or after genetic testing. Therefore, access to genetic testing and counseling is important for patient care. Health insurance coverage is a major determinant of access to health care in the United States. Uninsured individuals are less likely to have a regular source of health care than their insured counterparts. Different health insurance types and benefits also influence access to health care. Data on the association of health insurance and uptake of genetic testing and/or counseling for cancer risk are limited. Using data from the National Health Interview Survey, we examined the uptake of genetic testing and/or counseling for colorectal cancer (CRC) risk by health insurance type. We found that only a small proportion of individuals undergo genetic testing and/or counseling for CRC risk (0.8%), even among subgroups of individuals at risk due to family or personal history (3.7%). Insured individuals were more likely to undergo genetic testing and/or counseling for CRC risk, particularly those with Military and Private insurance plans, after adjusting for various demographic, socioeconomic, and health risk covariates. Further investigations are warranted to examine potential disparities in access and health inequities.
Collapse
Affiliation(s)
| | - Fang Zhang
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401 East, Boston, MA 02215, USA;
| | - Christine Y. Lu
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401 East, Boston, MA 02215, USA;
- Correspondence: ; Tel.: +1-617-867-4989
| |
Collapse
|
20
|
Uson PLS, Riegert-Johnson D, Boardman L, Kisiel J, Mountjoy L, Patel N, Lizaola-Mayo B, Borad MJ, Ahn D, Sonbol MB, Jones J, Leighton JA, Gurudu S, Singh H, Klint M, Kunze KL, Golafshar MA, Esplin ED, Nussbaum RL, Stewart AK, Bekaii-Saab TS, Jewel Samadder N. Germline Cancer Susceptibility Gene Testing in Unselected Patients With Colorectal Adenocarcinoma: A Multicenter Prospective Study. Clin Gastroenterol Hepatol 2022; 20:e508-e528. [PMID: 33857637 DOI: 10.1016/j.cgh.2021.04.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 04/03/2021] [Accepted: 04/06/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Hereditary factors play a role in the development of colorectal cancer (CRC). Identification of germline predisposition can have implications on treatment and cancer prevention. This study aimed to determine the prevalence of pathogenic germline variants (PGVs) in CRC patients using a universal testing approach, association with clinical outcomes, and the uptake of family variant testing. METHODS We performed a prospective multisite study of germline sequencing using a more than 80-gene next-generation sequencing platform among CRC patients (not selected for age or family history) receiving care at Mayo Clinic Cancer Centers between April 1, 2018, and March 31, 2020. RESULTS Of 361 patients, the median age was 57 years (SD, 12.4 y), 43.5% were female, 82% were white, and 38.2% had stage IV disease. PGVs were found in 15.5% (n = 56) of patients, including 44 in moderate- and high-penetrance cancer susceptibility genes. Thirty-four (9.4%) patients had incremental clinically actionable findings that would not have been detected by practice guideline criteria or a CRC-specific gene panel. Only younger age at diagnosis was associated with the presence of PGVs (odds ratio, 1.99; 95% CI, 1.12-3.56). After a median follow-up period of 20.7 months, no differences in overall survival were seen between those with or without a PGV (P = .2). Eleven percent of patients had modifications in their treatment based on genetic findings. Family cascade testing was low (16%). CONCLUSIONS Universal multigene panel testing in CRC was associated with a modest, but significant, detection of heritable mutations over guideline-based testing. One in 10 patients had changes in their management based on test results. Uptake of cascade family testing was low, which is a concerning observation that warrants further study.
Collapse
Affiliation(s)
| | - Douglas Riegert-Johnson
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Jacksonville, Florida; Department of Clinical Genomics, Mayo Clinic; Center for Individualized Medicine, Mayo Clinic
| | - Lisa Boardman
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - John Kisiel
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Neej Patel
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Phoenix, Arizona
| | - Blanca Lizaola-Mayo
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Phoenix, Arizona
| | - Mitesh J Borad
- Division of Hematology and Medical Oncology; Center for Individualized Medicine, Mayo Clinic
| | - Daniel Ahn
- Division of Hematology and Medical Oncology
| | | | - Jeremy Jones
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Jacksonville, Florida
| | - Jonathan A Leighton
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Phoenix, Arizona
| | - Suryakanth Gurudu
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Phoenix, Arizona
| | - Harminder Singh
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Katie L Kunze
- Department of Health Services Research, Mayo Clinic, Phoenix, Arizona
| | | | | | | | - A Keith Stewart
- Division of Hematology and Medical Oncology; Department of Clinical Genomics, Mayo Clinic; Center for Individualized Medicine, Mayo Clinic
| | | | - Niloy Jewel Samadder
- Department of Clinical Genomics, Mayo Clinic; Center for Individualized Medicine, Mayo Clinic; Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Phoenix, Arizona.
| |
Collapse
|
21
|
Long JM, Powers JM, Katona BW. Evaluation of Classic, Attenuated, and Oligopolyposis of the Colon. Gastrointest Endosc Clin N Am 2022; 32:95-112. [PMID: 34798989 PMCID: PMC8607742 DOI: 10.1016/j.giec.2021.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The goal of this review is to provide an overview of evaluating patients with adenomatous polyposis of the colon, including elements such as generating a differential diagnosis, referral considerations for genetic testing, genetic testing options, and expected outcomes from genetic testing in these individuals. In more recent years, adenomatous colonic polyposis has evolved beyond the more robustly characterized familial adenomatous polyposis (FAP) and MUTYH-associated polyposis (MAP) now encompassing more newly described genes and associated syndromes. Technological innovation, from whole-exome sequencing to multigene panel testing, has dramatically increased the amount of genotypic and phenotypic data amassed in adenomatous polyposis cohorts, which has contributed greatly to informing diagnosis and clinical management of affected individuals and their families.
Collapse
Affiliation(s)
- Jessica M. Long
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jacquelyn M. Powers
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Bryson W. Katona
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
22
|
Gupta N, Drogan C, Kupfer SS. How many is too many? Polyposis syndromes and what to do next. Curr Opin Gastroenterol 2022; 38:39-47. [PMID: 34839308 PMCID: PMC8648991 DOI: 10.1097/mog.0000000000000796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW The goal of this review is to help providers recognize, diagnose and manage gastrointestinal (GI) polyposis syndromes. RECENT FINDINGS Intestinal polyps include a number of histological sub-types such as adenomas, serrated, hamartomas among others. Over a quarter of individuals undergoing screening colonoscopy are expected to have colonic adenomas. Although it is not uncommon for adults to have a few GI polyps in their lifetime, some individuals are found to have multiple polyps of varying histology throughout the GI tract. In these individuals, depending on polyp histology, number, location and size as well as extra-intestinal features and/or family history, a polyposis syndrome should be considered with appropriate testing and management. SUMMARY Diagnosis and management of polyposis syndromes has evolved with advent of multigene panel testing and new data on optimal surveillance strategies. Evidence-based recommendations and current practice guidelines for polyposis syndromes are reviewed here. Areas of uncertainty and future research are also highlighted.
Collapse
Affiliation(s)
- Nina Gupta
- Section of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | | | | |
Collapse
|
23
|
Time to Classify Tumours of the Stomach and the Kidneys According to Cell of Origin. Int J Mol Sci 2021; 22:ijms222413386. [PMID: 34948181 PMCID: PMC8707540 DOI: 10.3390/ijms222413386] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 12/18/2022] Open
Abstract
Malignant tumours are traditionally classified according to their organ of origin and whether they are of epithelial (carcinomas) or mesenchymal (sarcomas) origin. By histological appearance the site of origin may often be confirmed. Using same treatment for tumours from the same organ is rational only when there is no principal heterogeneity between the tumours of that organ. Organ tumour heterogeneity is typical for the lungs with small cell and non-small cell tumours, for the kidneys where clear cell renal carcinoma (CCRCC) is the dominating type among other subgroups, and in the stomach with adenocarcinomas of intestinal and diffuse types. In addition, a separate type of neuroendocrine tumours (NETs) is found in most organs. Every cell type able to divide may develop into a tumour, and the different subtypes most often reflect different cell origin. In this article the focus is on the cells of origin in tumours arising in the stomach and kidneys and the close relationship between normal neuroendocrine cells and NETs. Furthermore, that the erythropoietin producing cell may be the cell of origin of CCRCC (a cancer with many similarities to NETs), and that gastric carcinomas of diffuse type may originate from the ECL cell, whereas the endodermal stem cell most probably gives rise to cancers of intestinal type.
Collapse
|
24
|
Tumor Classification Should Be Based on Biology and Not Consensus: Re-Defining Tumors Based on Biology May Accelerate Progress, An Experience of Gastric Cancer. Cancers (Basel) 2021; 13:cancers13133159. [PMID: 34202596 PMCID: PMC8269176 DOI: 10.3390/cancers13133159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/14/2021] [Accepted: 06/21/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Rational treatment of diseases including cancers depends on knowledge of their cause as well as their development. The present review is based upon more than 40 years’ work in clinical gastroenterology, gastric physiology, and pathology. The central role of hormones as well as local endocrine cells in cancer development has become apparent. Moreover, the classification of tumors should focus not only on the organ of origin but also on the cell of origin. All cells with the ability to divide may give rise to tumors. Based upon knowledge of the growth regulation of the cell of origin, prophylaxis and treatment may be tailored. Presently, there is hope for individual treatment of cancer patients based upon genetic analyses of tumors. However, with correct identification of the cell of origin, this may not be necessary. Abstract Malignant tumors are a consequence of genetic changes mainly occurring during cell division, sometimes with a congenital component. Therefore, accelerated cell divisions will necessarily predispose individuals, whether due to conditions of chronic cell destruction or hormonal overstimulation. It has been postulated that two genetic hits are necessary for the development of malignancy (Knudson). The correct view is probably that the number of genetic changes needed depends on the role the mutated genes have in proliferation and growth control. Hormones should accordingly be regarded as complete carcinogens. In this review based upon experience of gastric cancer where gastrin is central in the pathogenesis, it is argued that oxyntic atrophy—and not metaplasia as postulated by Correa—is the central precancer change in gastric mucosa. Moreover, the target cell of gastrin, the enterochromaffin-like (ECL) cell, is central in gastric carcinogenesis and most probably the cell of origin of gastric carcinomas of the diffuse type according to Lauren (a classification probable in accordance with biology). The distinction between adenocarcinomas and neuroendocrine carcinomas based upon a certain percentage of cancer cells with neuroendocrine differentiation is questioned. To make progress in the treatment of cancer, a correct classification system and knowledge of the pathogenesis are necessary.
Collapse
|
25
|
Anderson JL, Kruisselbrink TM, Lisi EC, Hughes TM, Steyermark JM, Winkler EM, Berg CM, Vierkant RA, Gupta R, Ali AH, Faubion SS, Aoudia SL, McAllister TM, Farrugia G, Stewart AK, Lazaridis KN. Clinically Actionable Findings Derived From Predictive Genomic Testing Offered in a Medical Practice Setting. Mayo Clin Proc 2021; 96:1407-1417. [PMID: 33890576 DOI: 10.1016/j.mayocp.2020.08.051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To assess the presence of clinically actionable results and other genetic findings in an otherwise healthy population of adults seen in a medical practice setting and offered "predictive" genomic testing. PATIENTS AND METHODS In 2014, a predictive genomics clinic for generally healthy adults was launched through the Mayo Clinic Executive Health Program. Self-identified interested patients met with a genomic nurse and genetic counselor for pretest advice and education. Two genome sequencing platforms and one gene panel-based health screen were offered. Posttest genetic counseling was available for patients who elected testing. From March 1, 2014, through June 1, 2019, 1281 patients were seen and 301 (23.5%) chose testing. Uptake rates increased to 36.3% [70 of 193]) in 2019 from 11.8% [2 of 17] in 2014. Clinically actionable results and genetic findings were analyzed using descriptive statistics. RESULTS Clinically actionable results were detected in 11.6% of patients (35 of 301), and of those, 51.7% (15 of 29) with a cancer or cardiovascular result = did not have a personal or family history concerning for a hereditary disorder. The most common actionable results were in the BCHE, BRCA2, CHEK2, LDLR, MUTYH, and MYH7 genes. A carrier of at least one recessive condition was found in 53.8% of patients (162 of 301). At least one variant associated with multifactorial disease was found in 44.5% (134 of 301) (eg, 25 patients were heterozygous for the F5 factor V Leiden variant associated with thrombophilia risk). CONCLUSION Our predictive screening revealed that 11.6% of individuals will test positive for a clinically actionable, likely pathogenic/pathogenic variant. This finding suggests that wider knowledge and adoption of predictive genomic services could be beneficial in medical practice, although additional studies are needed.
Collapse
Affiliation(s)
| | | | - Emily C Lisi
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | | | - Erin M Winkler
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Corinne M Berg
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Robert A Vierkant
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | - Ruchi Gupta
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | - Ahmad H Ali
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | | | - Stacy L Aoudia
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | - Gianrico Farrugia
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - A Keith Stewart
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Konstantinos N Lazaridis
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN; Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN.
| |
Collapse
|
26
|
Vanoli A, Grillo F, Furlan D, Arpa G, Grami O, Guerini C, Riboni R, Mastracci L, Di Sabatino A. Small Bowel Epithelial Precursor Lesions: A Focus on Molecular Alterations. Int J Mol Sci 2021; 22:ijms22094388. [PMID: 33922305 PMCID: PMC8122855 DOI: 10.3390/ijms22094388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/18/2021] [Accepted: 04/20/2021] [Indexed: 12/15/2022] Open
Abstract
The wider use of gastrointestinal endoscopic procedures has led to an increased detection of small intestinal preneoplastic and neoplastic epithelial lesions, most of which are identified in the duodenum and ampullary region. Like their malignant counterparts, small intestinal glandular precursor lesions, which include adenomas and hamartomas, may arise sporadically or be associated with hereditary tumor syndromes, such as familial adenomatous polyposis, MUTYH-associated polyposis, Lynch syndrome, Peutz-Jeghers syndrome, juvenile polyposis syndrome, and Cowden syndrome. In addition, dysplastic, preinvasive lesions have been observed adjacent to small bowel adenocarcinomas complicating immune-related disorders, such as celiac or Crohn’s disease. Adenomatous lesions may exhibit an intestinal-type, gastric-type, or, very rarely, serrated differentiation, related to different molecular pathogenetic mechanisms. Finally, in the background of multiple endocrine neoplasia 1 syndrome, precursor neuroendocrine growths have been described. In this review we offer a comprehensive description on the histo-molecular features of the main histotypes of small bowel epithelial precursors lesions, including: (i) sporadic adenomas (intestinal-type and gastric-type; non-ampullary and ampullary); (ii) syndromic adenomas; (iii) small bowel dysplasia in celiac and Crohn’s disease; (iv) serrated lesions; (v) hamartomatous lesions; and (vi) neuroendocrine precursor lesions.
Collapse
Affiliation(s)
- Alessandro Vanoli
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Lombardy, Italy; (G.A.); (O.G.); (C.G.); (R.R.)
- Correspondence: ; Tel.: +39-0382503612
| | - Federica Grillo
- Pathology Unit, Department of Surgical and Diagnostic Sciences, University of Genoa and Ospedale Policlinico San Martino University Hospital, 16132 Genoa, Liguria, Italy; (F.G.); (L.M.)
| | - Daniela Furlan
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, 21100 Varese, Lombardy, Italy;
| | - Giovanni Arpa
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Lombardy, Italy; (G.A.); (O.G.); (C.G.); (R.R.)
| | - Oneda Grami
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Lombardy, Italy; (G.A.); (O.G.); (C.G.); (R.R.)
| | - Camilla Guerini
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Lombardy, Italy; (G.A.); (O.G.); (C.G.); (R.R.)
| | - Roberta Riboni
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Lombardy, Italy; (G.A.); (O.G.); (C.G.); (R.R.)
| | - Luca Mastracci
- Pathology Unit, Department of Surgical and Diagnostic Sciences, University of Genoa and Ospedale Policlinico San Martino University Hospital, 16132 Genoa, Liguria, Italy; (F.G.); (L.M.)
| | - Antonio Di Sabatino
- Department of Internal Medicine, University of Pavia and Fondazione IRCCS San Matteo Hospital, 27100 Pavia, Lombardy, Italy;
| |
Collapse
|
27
|
Taylor H, Yerlioglu D, Phen C, Ballauff A, Nedelkopoulou N, Spier I, Loverdos I, Busoni VB, Heise J, Dale P, de Meij T, Sweet K, Cohen MC, Fox VL, Mas E, Aretz S, Eng C, Buderus S, Thomson M, Rojas I, Uhlig HH. mTOR inhibitors reduce enteropathy, intestinal bleeding and colectomy rate in patients with juvenile polyposis of infancy with PTEN-BMPR1A deletion. Hum Mol Genet 2021; 30:1273-1282. [PMID: 33822054 PMCID: PMC8804886 DOI: 10.1093/hmg/ddab094] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022] Open
Abstract
Ultra-rare genetic disorders can provide proof of concept for efficacy of targeted therapeutics and reveal pathogenic mechanisms relevant to more common conditions. Juvenile polyposis of infancy (JPI) is caused by microdeletions in chromosome 10 that result in haploinsufficiency of two tumor suppressor genes: phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and bone morphogenetic protein receptor type IA (BMPR1A). Loss of PTEN and BMPR1A results in a much more severe phenotype than deletion of either gene alone, with infantile onset pan-enteric polyposis and a high mortality rate. No effective pharmacological therapy exists. A multi-center cohort analysis was performed to characterize phenotype and investigate the therapeutic effect of mammalian target of rapamycin (mTOR) inhibition (adverse events, disease progression, time to colectomy and mortality) in patients with JPI. Among 25 JPI patients identified (mean age of onset 13 months), seven received mTOR inhibitors (everolimus, n = 2; or sirolimus, n = 5). Treatment with an mTOR inhibitor reduced the risk of colectomy (hazard ratio = 0.27, 95% confidence interval = 0.07-0.954, P = 0.042) and resulted in significant improvements in the serum albumin level (mean increase = 16.3 g/l, P = 0.0003) and hemoglobin (mean increase = 2.68 g/dl, P = 0.0077). Long-term mTOR inhibitor treatment was well tolerated over an accumulated follow-up time of 29.8 patient years. No serious adverse events were reported. Early therapy with mTOR inhibitors offers effective, pathway-specific and personalized treatment for patients with JPI. Inhibition of the phosphoinositol-3-kinase-AKT-mTOR pathway mitigates the detrimental synergistic effects of combined PTEN-BMPR1A deletion. This is the first effective pharmacological treatment identified for a hamartomatous polyposis syndrome.
Collapse
Affiliation(s)
- Henry Taylor
- Department of Surgery and Cancer, Imperial College London, London SW7 2BX, UK
| | - Dilay Yerlioglu
- Faculty of Medicine, Istanbul University, Istanbul, Fatih 34093, Turkey
| | - Claudia Phen
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Antje Ballauff
- Zentrum für Kinder- und Jugendmedizin Gastroenterology, HELIOS Klinikum Krefeld, Krefeld, Nordrhein-Westfalen 47805, Germany
| | - Natalia Nedelkopoulou
- Pediatric Gastroenterology, Sheffield Children's Hospital NHS Foundation Trust, Sheffield, Yorkshire S10 2TH, UK
| | - Isabel Spier
- Institute of Human Genetics, Medical Faculty, University of Bonn, Bonn, Nordrhein-Westfalen 53012, Germany.,National Centre for Hereditary Tumor Syndromes, University Hospital Bonn, Bonn, Nordrhein-Westfalen 53126, Germany
| | - Inés Loverdos
- Pediatric Gastroenterology Hepatology and Nutrition Unit, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Sabadell, Catalonia comunidad 08208, Spain
| | - Veronica B Busoni
- Pediatric Gastroenterology, Hepatology and Liver-Intestine Transplantation Division, Hospital Italiano de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Jürgen Heise
- Zentrum für Kinder- und Jugendmedizin Gastroenterology, HELIOS Klinikum Krefeld, Krefeld, Nordrhein-Westfalen 47805, Germany
| | - Peter Dale
- Royal Gwent Hospital, Newport NP20 2UB, UK
| | - Tim de Meij
- VU University Medical Center, Amsterdam 1081, The Netherlands
| | - Kevin Sweet
- Division of Human Genetics, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Marta C Cohen
- Histopathology Department, Sheffield Children's Hospital NHS Foundation Trust, Sheffield, Yorkshire S10 2TH, UK
| | - Victor L Fox
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA 02115, USA
| | - Emmanuel Mas
- Unité de Gastroentérologie, Hépatologie, Nutrition, Diabétologie et Maladies Héréditaires du Métabolisme, Hôpital des Enfants, CHU de Toulouse, and IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, Occitanie 31300, France
| | - Stefan Aretz
- Institute of Human Genetics, Medical Faculty, University of Bonn, Bonn, Nordrhein-Westfalen 53012, Germany.,National Centre for Hereditary Tumor Syndromes, University Hospital Bonn, Bonn, Nordrhein-Westfalen 53126, Germany
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, and Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Department of Genetics and Genome Sciences, and CASE Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Stephan Buderus
- GFO-Kliniken Bonn, St. Marien-Hospital, Bonn, Nordrhein-Westfalen 53115, Germany
| | - Mike Thomson
- Pediatric Gastroenterology, Sheffield Children's Hospital NHS Foundation Trust, Sheffield, Yorkshire S10 2TH, UK
| | - Isabel Rojas
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Holm H Uhlig
- Translational Gastroenterology Unit, University of Oxford, Oxford, Oxfordshire OX3 9DU, UK.,Department of Pediatrics, University of Oxford, Oxford, Oxfordshire OX3 9DU, UK.,Biomedical Research Centre, University of Oxford, Oxford, Oxfordshire OX4 2PG, UK
| |
Collapse
|
28
|
Abstract
Purpose of review Gastric cancer is a leading cause of cancer death in the world. Between 1% and 3% of cases are associated with specific genetic cancer risk syndromes. The purpose of this article is to review the latest insights, as well as gaps in knowledge, regarding some of the most common hereditary gastric cancer syndromes: hereditary diffuse gastric cancer (HDGC), gastric adenocarcinoma and proximal polyposis of the stomach (GAPPS), Lynch syndrome, the adenomatous polyposis syndromes, and the hamartomatous polyposis syndromes. Recent findings Patients carrying pathogenic variants in CDH1, but not meeting clinical criteria for HDGC, are increasingly being identified thanks to multigene panel testing; their absence from previous analyses overestimated gastric cancer penetrance. GAPPS is a recently described hereditary gastric cancer syndrome associated with specific point mutations in the promoter 1B region of the APC gene. Summary Risk of gastric cancer is highest among carriers of pathogenic variants in CDH1, with cumulative incidences approximately 40% and 30% for men and women, respectively. Mutations associated with Lynch syndrome and adenomatous polyposis syndromes confer greatest risk for gastric cancer in East Asian populations. Risk of gastric cancer in GAPPS and hamartomatous polyposis syndromes is difficult to estimate due to their rarity, but mutation status likely determines risk. Future research is needed to more precisely define risk of gastric cancer in these syndromes, so strategies for screening and prophylactic gastrectomy can be optimized.
Collapse
|
29
|
Samadder NJ, Riegert-Johnson D, Boardman L, Rhodes D, Wick M, Okuno S, Kunze KL, Golafshar M, Uson PLS, Mountjoy L, Ertz-Archambault N, Patel N, Rodriguez EA, Lizaola-Mayo B, Lehrer M, Thorpe CS, Yu NY, Esplin ED, Nussbaum RL, Sharp RR, Azevedo C, Klint M, Hager M, Macklin-Mantia S, Bryce AH, Bekaii-Saab TS, Sekulic A, Stewart AK. Comparison of Universal Genetic Testing vs Guideline-Directed Targeted Testing for Patients With Hereditary Cancer Syndrome. JAMA Oncol 2021; 7:230-237. [PMID: 33126242 PMCID: PMC7600058 DOI: 10.1001/jamaoncol.2020.6252] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/20/2020] [Indexed: 02/06/2023]
Abstract
IMPORTANCE Hereditary factors play a key role in the risk of developing several cancers. Identification of a germline predisposition can have important implications for treatment decisions, risk-reducing interventions, cancer screening, and germline testing. OBJECTIVE To examine the prevalence of pathogenic germline variants (PGVs) in patients with cancer using a universal testing approach compared with targeted testing based on clinical guidelines and the uptake of cascade family variant testing (FVT). DESIGN, SETTING, AND PARTICIPANTS This prospective, multicenter cohort study assessed germline genetic alterations among patients with solid tumor cancer receiving care at Mayo Clinic cancer centers and a community practice between April 1, 2018, and March 31, 2020. Patients were not selected based on cancer type, disease stage, family history of cancer, ethnicity, or age. EXPOSURES Germline sequencing using a greater than 80-gene next-generation sequencing platform. MAIN OUTCOMES AND MEASURES Proportion of PGVs detected with a universal strategy compared with a guideline-directed approach and uptake of cascade FVT in families. RESULTS A total of 2984 patients (mean [SD] age, 61.4 [12.2] years; 1582 [53.0%] male) were studied. Pathogenic germline variants were found in 397 patients (13.3%), including 282 moderate- and high-penetrance cancer susceptibility genes. Variants of uncertain significance were found in 1415 patients (47.4%). A total of 192 patients (6.4%) had incremental clinically actionable findings that would not have been detected by phenotype or family history-based testing criteria. Of those with a high-penetrance PGV, 42 patients (28.2%) had modifications in their treatment based on the finding. Only younger age of diagnosis was associated with presence of PGV. Only 70 patients (17.6%) with PGVs had family members undergoing no-cost cascade FVT. CONCLUSIONS AND RELEVANCE This prospective, multicenter cohort study found that universal multigene panel testing among patients with solid tumor cancer was associated with an increased detection of heritable variants over the predicted yield of targeted testing based on guidelines. Nearly 30% of patients with high-penetrance variants had modifications in their treatment. Uptake of cascade FVT was low despite being offered at no cost.
Collapse
Affiliation(s)
- N. Jewel Samadder
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Phoenix, Arizona
- Department of Clinical Genomics, Mayo Clinic, Phoenix, Arizona
- Center for Individualized Medicine, Mayo Clinic, Phoenix, Arizona
| | - Douglas Riegert-Johnson
- Department of Clinical Genomics, Mayo Clinic, Phoenix, Arizona
- Center for Individualized Medicine, Mayo Clinic, Phoenix, Arizona
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic Florida, Jacksonville
| | - Lisa Boardman
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, New York
| | - Deborah Rhodes
- Department of Medicine, Yale New Haven Hospital, New Haven, Connecticut
- Formerly with Division of General Internal Medicine, Department of Medicine, Mayo Clinic Rochester, New York
| | - Myra Wick
- Department of Clinical Genomics, Mayo Clinic, Phoenix, Arizona
- Center for Individualized Medicine, Mayo Clinic, Phoenix, Arizona
- Department of Obstetrics and Gynecology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Scott Okuno
- Department of Hematology and Oncology, Mayo Clinic Health System, Eau Claire, Wisconsin
| | - Katie L. Kunze
- Department of Health Sciences Research, Mayo Clinic Arizona, Phoenix
| | - Michael Golafshar
- Department of Health Sciences Research, Mayo Clinic Arizona, Phoenix
| | - Pedro L. S. Uson
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic Arizona, Phoenix
| | - Luke Mountjoy
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic Arizona, Phoenix
| | - Natalie Ertz-Archambault
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic Arizona, Phoenix
| | - Neej Patel
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Phoenix, Arizona
| | - Eduardo A. Rodriguez
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Phoenix, Arizona
| | - Blanca Lizaola-Mayo
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Phoenix, Arizona
| | | | | | - Nathan Y. Yu
- Department of Radiation Oncology, Mayo Clinic Arizona, Phoenix
| | | | | | - Richard R. Sharp
- Center for Individualized Medicine, Mayo Clinic, Phoenix, Arizona
- Department of Bioethics, Mayo Clinic, Rochester, Minnesota
| | - Cindy Azevedo
- Center for Individualized Medicine, Mayo Clinic, Phoenix, Arizona
| | - Margaret Klint
- Department of Clinical Genomics, Mayo Clinic, Phoenix, Arizona
| | - Megan Hager
- Department of Clinical Genomics, Mayo Clinic, Phoenix, Arizona
| | | | - Alan H. Bryce
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic Arizona, Phoenix
| | - Tanios S. Bekaii-Saab
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic Arizona, Phoenix
| | - Aleksandar Sekulic
- Center for Individualized Medicine, Mayo Clinic, Phoenix, Arizona
- Department of Dermatology, Mayo Clinic Arizona, Phoenix
| | - A. Keith Stewart
- Center for Individualized Medicine, Mayo Clinic, Phoenix, Arizona
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic Arizona, Phoenix
| |
Collapse
|
30
|
Kővári B, Kim BH, Lauwers GY. The pathology of gastric and duodenal polyps: current concepts. Histopathology 2020; 78:106-124. [PMID: 33382489 DOI: 10.1111/his.14275] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/25/2020] [Accepted: 10/01/2020] [Indexed: 12/26/2022]
Abstract
The liberal use of upper endoscopy has led to an increased detection of gastric and duodenal polyps, which are identified in as many as 6 and 4.6% of patient examinations, respectively. Gastroduodenal polyps are a heterogeneous group of lesions that can be neoplastic or non-neoplastic (e.g. hyperplastic or heterotopical). Most polyps present characteristic topographical features, as well as endoscopic appearance and size. Evaluation of the surrounding mucosa is essential in assessing the underlying pathology (e.g. Helicobacter pylori, autoimmune gastritis or inherited polyposis syndromes). Phylogenetically, gastric and duodenal polyps can be classified according to the epithelial compartment from which they derive. Polyps that arise from the surface epithelium can either be of foveolar or intestinal type, and they can develop from either the native mucosa or the metaplastic epithelium (gastric intestinal metaplasia or duodenal foveolar metaplasia). Other polyps develop from the deeper glandular component, such as pyloric/oxyntic gland derived subtypes. In this review we focus upon epithelial polyps, with an emphasis on the most common and clinically relevant lesions, and present recently described entities.
Collapse
Affiliation(s)
- Bence Kővári
- Department of Pathology, University of Szeged and Albert Szent-Györgyi Health Center, Szeged, Hungary.,Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute and Departments of Pathology and Oncologic Sciences, University of South Florida, Tampa, FL, USA
| | - Baek H Kim
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute and Departments of Pathology and Oncologic Sciences, University of South Florida, Tampa, FL, USA.,Department of Pathology, Korea University Guro Hospital, Seoul, Korea
| | - Gregory Y Lauwers
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute and Departments of Pathology and Oncologic Sciences, University of South Florida, Tampa, FL, USA
| |
Collapse
|
31
|
Akcay IM, Celik E, Agaoglu NB, Alkurt G, Kizilboga Akgun T, Yildiz J, Enc F, Kir G, Canbek S, Kilic A, Zemheri E, Ezberci F, Ozcelik M, Dinler Doganay G, Doganay L. Germline pathogenic variant spectrum in 25 cancer susceptibility genes in Turkish breast and colorectal cancer patients and elderly controls. Int J Cancer 2020; 148:285-295. [PMID: 32658311 DOI: 10.1002/ijc.33199] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/21/2020] [Accepted: 06/23/2020] [Indexed: 12/19/2022]
Abstract
Inherited pathogenic variants account for 5% to 10% of all breast cancer (BC) and colorectal cancer (CRC) cases. Here, we sought to profile the pathogenic variants in 25 cancer susceptibility genes in Turkish population. Germline pathogenic variants were screened in 732 BC patients, 189 CRC patients and 490 cancer-free elderly controls, using next-generation sequencing-based multigene panel testing and multiplex ligation-dependent probe amplification testing. Pathogenic variants were detected in 17.2% of high-risk BC patients and 26.4% of high-risk CRC patients. More than 95% of these variants were clinically actionable. BRCA1/2 and mismatch repair genes (MLH1, MSH2 and MSH6) accounted for two-thirds of all pathogenic variants detected in high-risk BC and CRC patients, respectively. Pathogenic variants in PALB2, CHEK2, ATM and TP53 were also prevalent in high-risk BC patients (4.5%). BRCA1 exons 17-18 deletion and CHEK2 c.592+3A>T were the most common variants predisposing to BC, and they are likely to be founder variants. Three frequent MUTYH pathogenic variants (c.884C>T, c.1437_1439delGGA and c.1187G>A) were responsible for all MUTYH biallelic cases (4.4% of high-risk CRC patients). The total pathogenic variant frequency was very low in controls (2.4%) and in low-risk BC (3.9%) and CRC (6.1%) patients. Our study depicts the pathogenic variant spectrum and prevalence in Turkish BC and CRC patients, guiding clinicians and health authorities for genetic testing applications and variant classification in Turkish population.
Collapse
Affiliation(s)
- Izzet Mehmet Akcay
- Department of Molecular Biology-Genetics & Biotechnology, Istanbul Technical University, Istanbul, Turkey.,GLAB (Genomic Laboratory), Health Directorate of Istanbul, Istanbul, Turkey
| | - Elifnaz Celik
- Department of Molecular Biology-Genetics & Biotechnology, Istanbul Technical University, Istanbul, Turkey.,GLAB (Genomic Laboratory), Health Directorate of Istanbul, Istanbul, Turkey
| | - Nihat Bugra Agaoglu
- GLAB (Genomic Laboratory), Health Directorate of Istanbul, Istanbul, Turkey.,Department of Clinical Genetics, Umraniye Teaching and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Gizem Alkurt
- Department of Molecular Biology-Genetics & Biotechnology, Istanbul Technical University, Istanbul, Turkey.,GLAB (Genomic Laboratory), Health Directorate of Istanbul, Istanbul, Turkey
| | - Tugba Kizilboga Akgun
- Department of Molecular Biology-Genetics & Biotechnology, Istanbul Technical University, Istanbul, Turkey.,GLAB (Genomic Laboratory), Health Directorate of Istanbul, Istanbul, Turkey
| | - Jale Yildiz
- Department of Molecular Biology-Genetics & Biotechnology, Istanbul Technical University, Istanbul, Turkey.,GLAB (Genomic Laboratory), Health Directorate of Istanbul, Istanbul, Turkey
| | - Feruze Enc
- Department of Gastroenterology, Goztepe Teaching and Research Hospital, Medeniyet University, Istanbul, Turkey
| | - Gozde Kir
- Department of Pathology, Goztepe Teaching and Research Hospital, Medeniyet University, Istanbul, Turkey
| | - Sezin Canbek
- GLAB (Genomic Laboratory), Health Directorate of Istanbul, Istanbul, Turkey.,Department of Clinical Genetics, Umraniye Teaching and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Ali Kilic
- Department of General Surgery, Umraniye Teaching and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Ebru Zemheri
- Department of Pathology, Umraniye Teaching and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Fikret Ezberci
- Department of General Surgery, Umraniye Teaching and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Melike Ozcelik
- Department of Oncology, Kartal Lutfi Kirdar Teaching and Research Hospital, University of Health Sciences, Istanbul, Turkey.,Department of Oncology, Umraniye Teaching and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Gizem Dinler Doganay
- Department of Molecular Biology-Genetics & Biotechnology, Istanbul Technical University, Istanbul, Turkey.,GLAB (Genomic Laboratory), Health Directorate of Istanbul, Istanbul, Turkey
| | - Levent Doganay
- GLAB (Genomic Laboratory), Health Directorate of Istanbul, Istanbul, Turkey.,Department of Gastroenterology and Hepatology, Umraniye Teaching and Research Hospital, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
32
|
Sikorska A, Stachowiak M, Flisikowska T, Stachecka J, Flisikowski K, Switonski M. Polymorphisms of CSF1R and WISP1 genes are associated with severity of familial adenomatous polyposis in APC 1311 pigs. Gene 2020; 759:144988. [PMID: 32717306 DOI: 10.1016/j.gene.2020.144988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/01/2020] [Accepted: 07/21/2020] [Indexed: 12/24/2022]
Abstract
Hereditary familial adenomatous polyposis (FAP) in humans significantly increases the risk of development of colorectal cancer (CRC). Germline mutations in the APC (adenomatous polyposis coli) gene are responsible for FAP. Despite having the same causative mutation, the severity of the disease differs from patient to patient. The porcine FAP model carrying a truncating APC1311 mutation, orthologous to the dominant human mutation that leads to severe form of the disease (APC1309), mirrors the severity of polyposis. Earlier RNAseq studies have revealed the differential expression of WISP1 and CSF1R in samples derived from low-grade (LG-IEN) and more advanced high-grade (HG-IEN) colon polyps of APC1311/+ pigs. The grade of dysplasia was correlated with the severity of polyposis in APC1311/+ pigs characterized by a low (LP) and high (HP) numbers of polyps. The goal of this work was to find DNA variants that regulate the expression of CSF1R and WISP1 in LP and HP pigs. In total, 32 and 36 polymorphisms in CSF1R and WISP1 were found, respectively. Of these, the genotype frequency of four silent SNPs in the coding region of WISP1 differed significantly between LP and HP lines. In silico analysis revealed an elevated minimum free energy (MFE) for three of these SNPs, suggesting their role in mRNA structure stability. Furthermore, four polymorphisms in the promoter region of CSF1R, cosegregating as a common haplotype, were associated with polyp number in APC1311/+ pigs. A secreted alkaline phosphatase (SEAP) assay showed, however, that these variants have no direct effect on the activity of the CSF1R promoter. Concluding, our study identified polymorphisms in CSF1R and WISP1 that are potentially associated with the severity of polyposis in APC1311/+ pigs.
Collapse
Affiliation(s)
- Agata Sikorska
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Wolynska 33, 60-637 Poznan, Poland
| | - Monika Stachowiak
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Wolynska 33, 60-637 Poznan, Poland
| | - Tatiana Flisikowska
- Chair of Livestock Biotechnology, Technical University of Munich, Liesel-Beckmannstr. 1, 85354 Freising, Germany
| | - Joanna Stachecka
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Wolynska 33, 60-637 Poznan, Poland
| | - Krzysztof Flisikowski
- Chair of Livestock Biotechnology, Technical University of Munich, Liesel-Beckmannstr. 1, 85354 Freising, Germany.
| | - Marek Switonski
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Wolynska 33, 60-637 Poznan, Poland.
| |
Collapse
|
33
|
Gatalica Z, Vranic S, Krušlin B, Poorman K, Stafford P, Kacerovska D, Senarathne W, Florento E, Contreras E, Leary A, Choi A, In GK. Comparison of the biomarkers for targeted therapies in primary extra-mammary and mammary Paget's disease. Cancer Med 2020; 9:1441-1450. [PMID: 31899853 PMCID: PMC7013075 DOI: 10.1002/cam4.2820] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 12/14/2022] Open
Abstract
Background Primary Extra‐mammary Paget's disease (EMPD) is a very rare cutaneous adenocarcinoma affecting anogenital or axillary regions. It is characterized by a prolonged course with recurrences and eventually distant metastatic spread for which no specific therapy is known. Methods Eighteen EMPD (13 vulvar and five scrotal) and ten mammary Paget's disease (MPD) cases were comprehensively profiled for gene mutations, fusions and copy number alterations, and for therapy‐relevant protein biomarkers). Results Mutations in TP53 and PIK3CA were the most frequent in both cohorts: 7/15 and 5/15 in EMPD; 1/6 and 4/7 in MPD HER2 gene amplification was detected in 4/18 EMPD (3 vulvar and 1 scrotal case) in contrast to MPD where it was detected in the majority (7/8) of cases. TOP2A gene amplification was seen in 2/12 EMPD and 1/6 MPD, respectively. Similarly, no difference in estrogen receptor expression was seen between the EMPD (4/15) and MPD (3/10). Androgen receptor was also expressed in the majority of both cohorts (12/16 EMPD) and (7/8 MPD).Here ARv7 splice variant was detected in 1/7 EMPD and 1/4 MPD cases, respectively. PD‐L1 expression on immune cells was exclusively observed in three vulvar EMPD. In contrast to MPD, six EMPDs harbored a “high” tumor mutation burden (≥10 mutations/Mb). All tested cases from both cohorts were MSI stable. Conclusions EMPD shares some targetable biomarkers with its mammary counterpart (steroid receptors, PIK3CA signaling pathways, TOP2A amplification). HER2 positivity is notably lower in EMPD while biomarkers to immune checkpoint inhibitors (high TMB and PD‐L1) were observed in some EMPD. Given that no consistent molecular alteration characterizes EMPD, comprehensive theranostic profiling is required to identify individual patients with targetable molecular alterations.
Collapse
Affiliation(s)
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Božo Krušlin
- Ljudevit Jurak Department of Pathology and Cytology, Clinical Hospital Center Sestre Milosrdnice, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | | | | | - Denisa Kacerovska
- Medical Faculty in Pilsen, Sikl's Department of Pathology, Charles University in Prague, Pilsen, Czech Republic.,Bioptical Laboratory, Pilsen, Czech Republic
| | | | | | | | | | - April Choi
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Gino K In
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Colorectal cancer (CRC) is the third most common cancer in the USA and inherited cancer syndromes are responsible for approximately 3-5% of all CRCs. Genetic testing costs have plummeted in recent years; however, awareness and referral of high-risk patients for testing is still very low. We review the salient clinical features, genetics, and management of well-defined gastrointestinal (GI) hereditary polyposis syndromes including familial adenomatous polyposis, MUTYH-associated polyposis, and the hamartomatous polyposis syndromes. RECENT FINDINGS Comprehensive endoscopic surveillance has the potential to prevent the development of GI cancer and to identify early-stage cancer; newer developments like high-definition endoscopes, chromoendoscopy, and the use of cap-assisted endoscopy have shown promise for enhanced lesion detection rates. Several chemoprevention trials have yielded promising results but safety and efficacy data for long-term use is still awaited. Several new polyposis genes have also been identified in the recent years. Multiple societies have recently published updated surveillance guidelines to aid clinicians in the detection and management of patients with hereditary GI polyposis syndromes. Although these syndromes are rare, it is crucial for the clinicians to recognize these in a timely manner, for the appropriate management plans for both the patient and their at risk family members.
Collapse
|
35
|
Gupta S, Provenzale D, Llor X, Halverson AL, Grady W, Chung DC, Haraldsdottir S, Markowitz AJ, Slavin Jr TP, Hampel H, Ness RM, Weiss JM, Ahnen DJ, Chen LM, Cooper G, Early DS, Giardiello FM, Hall MJ, Hamilton SR, Kanth P, Klapman JB, Lazenby AJ, Lynch PM, Mayer RJ, Mikkelson J, Peter S, Regenbogen SE, Dwyer MA, Ogba N. NCCN Guidelines Insights: Genetic/Familial High-Risk Assessment: Colorectal, Version 2.2019. J Natl Compr Canc Netw 2019; 17:1032-1041. [DOI: 10.6004/jnccn.2019.0044] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Identifying individuals with hereditary syndromes allows for improved cancer surveillance, risk reduction, and optimized management. Establishing criteria for assessment allows for the identification of individuals who are carriers of pathogenic genetic variants. The NCCN Guidelines for Genetic/Familial High-Risk Assessment: Colorectal provide recommendations for the assessment and management of patients with high-risk colorectal cancer syndromes. These NCCN Guidelines Insights focus on criteria for the evaluation of Lynch syndrome and considerations for use of multigene testing in the assessment of hereditary colorectal cancer syndromes.
Collapse
Affiliation(s)
| | | | | | - Amy L. Halverson
- 4Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | - William Grady
- 5Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | | | | | | | | | | | | | | | - Lee-may Chen
- 14UCSF Helen Diller Family Comprehensive Cancer Center
| | - Gregory Cooper
- 15Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | - Dayna S. Early
- 16Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|