1
|
Wegert J, Appenzeller S, Treger TD, Streitenberger H, Ziegler B, Bausenwein S, Vokuhl C, Parks C, Jüttner E, Gramlich S, Ernestus K, Warman SW, Fuchs J, Hubertus J, von Schweinitz D, Fröhlich B, Jorch N, Knöfler R, Friedrich C, Corbacioglu S, Frühwald MC, Pekrun A, Schneider DT, Faber J, Stursberg J, Metzler M, Welter N, Pritchard-Jones K, Graf N, Furtwängler R, Behjati S, Gessler M. Distinct pathways for genetic and epigenetic predisposition in familial and bilateral Wilms tumor. Genome Med 2025; 17:49. [PMID: 40340749 PMCID: PMC12060375 DOI: 10.1186/s13073-025-01482-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 04/28/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Genetic predisposition is particularly common in children with the kidney cancer, Wilms tumor. In 10% of these children, this manifests as a family history of Wilms tumor or bilateral disease. The frequency and spectrum of underlying changes have not been systematically investigated. METHODS We analyzed 129 children with suspected Wilms tumor predisposition, 20 familial cases, and 109 children with bilateral disease, enrolled over 30 years in the German SIOP93-01/GPOH and SIOP2001 studies. We used whole exome, whole genome, and targeted DNA sequencing, together with MLPA and targeted methylation assays on tumor, blood, and normal kidney to determine predisposing changes. RESULTS Predisposing variants were identified in 117/129 children, comprising DNA variants (57%) and epigenetic changes (34%). Most children had predisposition variants in genes previously implicated in Wilms tumor: most prominently WT1 (n = 35) and less frequently TRIM28, REST, DIS3L2, CTR9, DICER1, CDC73, and NONO. Nine children carried germline mutations in cancer predisposition genes not considered Wilms tumor predisposition genes, such as CHEK2, CDKN2A, BLM, BRCA2, STK11, and FMN2. Predisposition via epigenetic BWS-IC1 alterations occurred as early somatic events, reflected by partial (mosaic) loss of imprinting or loss of heterozygosity at the IGF2/H19 locus in normal kidney or blood. These patients rarely had a clinical diagnosis of Beckwith-Wiedemann syndrome (BWS). Especially WT1-driven tumors follow a stereotypical pathway of germline WT1 mutations becoming homozygous in renal precursor lesions through 11p LOH, which concomitantly activates imprinted IGF2 expression, with subsequent WNT pathway activation leading to tumor growth. There is a high rate of multicentric tumors, which may have previously been missed in unilateral tumors. While Wilms tumor predisposition genes relied on somatic inactivation of the second allele, this was different for general cancer predisposition genes. The latter cases were often associated with additional oncogenic alterations, similar to tumors with epigenetic predisposition. CONCLUSIONS We identified two main mechanisms of Wilms tumor predisposition: either germline genetic alterations of Wilms tumor and, less frequently, general cancer genes; or postzygotic mosaic imprinting defects activating IGF2. These findings inform future genetic screening and risk assessment of affected children and lend support to liquid biopsy screening for enhanced therapeutic stratification.
Collapse
Affiliation(s)
- Jenny Wegert
- Developmental Biochemistry, Theodor-Boveri-Institute/Biocenter, Julius-Maximilians-University Würzburg, Am Hubland, Würzburg, 97074, Germany
| | - Silke Appenzeller
- Comprehensive Cancer Center Mainfranken, University Hospital of Würzburg, Würzburg, Germany
| | - Taryn D Treger
- Wellcome Sanger Institute, Hinxton, UK
- Department of Pediatrics, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Heike Streitenberger
- Developmental Biochemistry, Theodor-Boveri-Institute/Biocenter, Julius-Maximilians-University Würzburg, Am Hubland, Würzburg, 97074, Germany
| | - Barbara Ziegler
- Developmental Biochemistry, Theodor-Boveri-Institute/Biocenter, Julius-Maximilians-University Würzburg, Am Hubland, Würzburg, 97074, Germany
| | - Sabrina Bausenwein
- Developmental Biochemistry, Theodor-Boveri-Institute/Biocenter, Julius-Maximilians-University Würzburg, Am Hubland, Würzburg, 97074, Germany
| | - Christian Vokuhl
- Section of Pediatric Pathology, Department of Pathology, University Hospital Bonn, Bonn, Germany
| | | | - Eva Jüttner
- Department of Pathology, Schleswig-Holstein University Hospital, Kiel, Germany
| | - Susanne Gramlich
- Department of Pathology, University of Würzburg, Würzburg, Germany
| | - Karen Ernestus
- Department of Pathology, University of Würzburg, Würzburg, Germany
| | - Steven W Warman
- Clinic of Pediatric Surgery, Charité - University Hospital Berlin, Berlin, Germany
| | - Jörg Fuchs
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital, Tuebingen, Germany
| | - Jochen Hubertus
- Department of Pediatric Surgery, Marien Hospital Witten, Ruhr-University Bochum, Bochum, Germany
| | - Dietrich von Schweinitz
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, LMU University Hospital, Munich, Germany
| | - Birgit Fröhlich
- Department of Pediatric Oncology and Hematology, University of Münster, Münster, Germany
| | - Norbert Jorch
- Evangelisches Klinikum Bethel, Universitätsklinikum OWL, Bielefeld, Germany
| | - Ralf Knöfler
- Department of Pediatric Hematology/Oncology, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Carsten Friedrich
- Department of Pediatrics and Pediatric Hematology/Oncology, University Children's Hospital, Carl von Ossietzky University, Klinikum Oldenburg, Oldenburg, Germany
| | - Selim Corbacioglu
- Children's Hospital Regensburg, University of Regensburg, Regensburg, Germany
| | - Michael C Frühwald
- Swabian Children's Cancer Center, Pediatrics and Adolescent Medicine, University Hospital Augsburg, Augsburg, Germany
| | - Arnulf Pekrun
- Pediatric Hematology and Oncology, Klinikum Bremen, Bremen, Germany
| | - Dominik T Schneider
- Clinic of Pediatrics, University Witten/Herdecke, Klinikum Dortmund, Witten, Germany
| | - Jörg Faber
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Jana Stursberg
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Markus Metzler
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Nils Welter
- Department of Pediatric Hematology and Oncology, Saarland University Hospital, Homburg, Germany
| | - Kathy Pritchard-Jones
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Norbert Graf
- Department of Pediatric Hematology and Oncology, Saarland University Hospital, Homburg, Germany
| | - Rhoikos Furtwängler
- Department of Pediatric Hematology and Oncology, Saarland University Hospital, Homburg, Germany
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Inselspital University Hospital, University of Bern, Bern, Switzerland
| | - Sam Behjati
- Wellcome Sanger Institute, Hinxton, UK
- Department of Pediatrics, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Manfred Gessler
- Developmental Biochemistry, Theodor-Boveri-Institute/Biocenter, Julius-Maximilians-University Würzburg, Am Hubland, Würzburg, 97074, Germany.
- Comprehensive Cancer Center Mainfranken, University Hospital of Würzburg, Würzburg, Germany.
| |
Collapse
|
2
|
Tasdogan A, Sullivan RJ, Katalinic A, Lebbe C, Whitaker D, Puig S, van de Poll-Franse LV, Massi D, Schadendorf D. Cutaneous melanoma. Nat Rev Dis Primers 2025; 11:23. [PMID: 40180935 DOI: 10.1038/s41572-025-00603-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 04/05/2025]
Abstract
Cutaneous melanoma is a common cancer in Australia and New Zealand, Europe, and North America, and its incidence is still increasing in many regions. Ultraviolet (UV) radiation exposure (for example, through excessive sunlight exposure) remains the primary risk factor for melanoma; however, public awareness campaigns have led to a marked reduction in mortality. In addition to genetic damage from UV radiation, specific genetic alterations have been linked to melanoma. The stage of the tumour at the time of diagnosis is of greater importance for melanoma prognosis than in almost any other cancer. Context-dependent genetic mutations that attenuate tumour-suppressive mechanisms or activate growth-promoting signalling pathways are crucial factors in the development of cutaneous melanoma. In addition to external factors such as UV radiation, the tumour microenvironment can contribute to melanoma progression, invasion and metastasis. Cutaneous melanoma treatment has improved considerably over the past decade with the discovery and development of immune checkpoint inhibitors and therapy targeting BRAF and MEK. Over the next decade, several priorities are likely to influence melanoma research and management, including the continued advance of precision medicine methods to identify the most suitable patients for the most effective treatment, with the aim of improving clinical outcomes.
Collapse
Affiliation(s)
- Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen & German Cancer Consortium (DKTK), Partner Site Essen, Essen, Germany.
- National Center for Tumour diseases (NCT-West), Campus Essen & Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany.
| | - Ryan J Sullivan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Alexander Katalinic
- Institute for Social Medicine and Epidemiology, University of Lübeck, Lübeck, Germany
| | - Celeste Lebbe
- Université Paris Cite, AP-HP Dermato-oncology and CIC, Cancer institute APHP.nord Paris cité, INSERM U976, Saint Louis Hospital, Paris, France
| | - Dagmar Whitaker
- Melanoma Advisory Board South Africa, Cape Town, South Africa
| | - Susana Puig
- Dermatology Department, IDIBAPS, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
- 8CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Lonneke V van de Poll-Franse
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
- Department of Medical and Clinical Psychology, CoRPS - Center of Research on Psychology in Somatic Diseases, Tilburg University, Tilburg, Netherlands
| | - Daniela Massi
- Section of Pathology, Department of Health Sciences, University of Florence, Florence, Italy
- Department of Molecular Pathobiology, New York University - College of Dentistry, New York, NY, USA
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen & German Cancer Consortium (DKTK), Partner Site Essen, Essen, Germany.
- National Center for Tumour diseases (NCT-West), Campus Essen & Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
3
|
Shah Y, Dahiya DS, Tiwari A, Kumar H, Gangwani MK, Ali H, Hayat U, Alsakarneh S, Singh S, Malik S, Sohail AH, Chandan S, Ali MA, Inamdar S. Advancements in Early Detection and Screening Strategies for Pancreatic Cancer: From Genetic Susceptibility to Novel Biomarkers. J Clin Med 2024; 13:4706. [PMID: 39200847 PMCID: PMC11355237 DOI: 10.3390/jcm13164706] [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: 07/25/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
Pancreatic cancer is a rare but lethal cancer due to its biologically aggressive nature, advanced stage at the time of diagnosis, and poor response to oncologic therapies. The risk of pancreatic cancer is significantly higher to 5% in certain high-risk individuals with inherited genetic susceptibility. Screening for pancreatic cancer in these individuals from high-risk groups can help with the early detection of pancreatic cancer as well as the detection of precursor lesions leading to early surgical resection and improved overall outcomes. The advancements in radiological imaging as well as advanced endoscopic procedures has made a significant impact on the early diagnosis, surveillance, and staging of pancreatic cancer. There is also a significant advancement in the development of biomarkers for the early detection of pancreatic cancer, which has also led to the development of liquid biopsy, allowing for microRNA detection in serum and circulating tumor cells. Various societies and organizations have provided guidelines for pancreatic cancer screening and surveillance in high-risk individuals. In this review, we aim to discuss the hereditary risk factors for developing pancreatic cancer, summarize the screening recommendations by different societies, and discuss the development of novel biomarkers and areas for future research in pancreatic cancer screening for high-risk individuals.
Collapse
Affiliation(s)
- Yash Shah
- Department of Internal Medicine, Trinity Health Oakland/Wayne State University, Pontiac, MI 48341, USA
| | - Dushyant Singh Dahiya
- Division of Gastroenterology, Hepatology & Motility, The University of Kansas School of Medicine, Kansas City, KS 66160, USA
| | - Angad Tiwari
- Department of Internal Medicine, Maharani Laxmi Bai Medical College, Jhansi 284001, Uttar Pradesh, India
| | - Harendra Kumar
- Department of Internal Medicine, Dow University of Health Sciences, Karachi 74200, Pakistan
| | - Manesh Kumar Gangwani
- Department of Gastroenterology and Hepatology, University of Arkansas For Medical Sciences, Little Rock, AR 72205, USA
| | - Hassam Ali
- Division of Gastroenterology, Hepatology & Nutrition, East Carolina University/Brody School of Medicine, Greenville, NC 27834, USA
| | - Umar Hayat
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes Barre, PA 18711, USA
| | - Saqr Alsakarneh
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Sahib Singh
- Department of Internal Medicine, Sinai Hospital, Baltimore, MD 21215, USA
| | - Sheza Malik
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY 14621, USA
| | - Amir H. Sohail
- Department of Surgery, University of New Mexico, Albuquerque, NM 87131, USA
| | - Saurabh Chandan
- Center for Interventional Endoscopy (CIE), Advent Health, Orlando, FL 32803, USA
| | - Meer A. Ali
- Department of Gastroenterology and Hepatology, University of Arkansas For Medical Sciences, Little Rock, AR 72205, USA
| | - Sumant Inamdar
- Department of Gastroenterology and Hepatology, University of Arkansas For Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
4
|
Ferrara G, Paiella S, Settanni G, Frizziero M, Rosina P, Viassolo V. Prevalence of CDKN2A, CDK4, POT1, BAP1, MITF, ATM, and TERT Pathogenic Variants in a Single-Center Retrospective Series of Patients With Melanoma and Personal or Family History Suggestive of Genetic Predisposition. Dermatol Pract Concept 2024; 14:e2024120. [PMID: 39122510 PMCID: PMC11314473 DOI: 10.5826/dpc.1403a120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2024] [Indexed: 08/12/2024] Open
Abstract
INTRODUCTION Approximately 20%-45% of familial melanoma (FM) cases are associated with genetic predisposition. OBJECTIVES This single-center retrospective study aimed to assess the frequency of pathogenic variants (PV) in the main melanoma-predisposing genes in patients with cutaneous melanoma and investigate the clinical predictors of genetic predisposition. METHODS Patients included were those diagnosed with cutaneous melanoma at the Dermatology Unit of the University Hospital of Verona, Italy, from 2000 to 2022, presenting at least one of the followings: multiple melanomas (≥ 3); personal/family history of pancreatic cancer (PC) (up to 2nd-degree relatives); ≥ 2 1st-degree relatives with melanoma; ≥ 1 1st-degree relatives with early-onset (<45 years) melanoma and tested for CDKN2A, CDK4, POT1, BAP1, MITF, ATM, and TERT. RESULTS During the study period, 35 out of 1320 patients (2.7%) underwent genetic testing. Four patients (11.4%) harbored a PV in a melanoma-predisposing gene, three in CDKN2A (8.6%), and one in MITF (2.9%). Variants currently classified as being of unknown clinical significance (VUS) were detected in CDKN2A (N = 1), MITF (N = 1), and ATM (N = 2). Family history of PC and ≥5 melanomas, personal history of ≥50 nevi, and ≥4 melanomas were significantly associated with PV in tested genes (P < 0.05). CONCLUSIONS The prevalence of PV in predisposing genes in FM was lower than previously reported in Italian registries. Possible reasons include deleterious variants in untested intermediate/low-penetrance genes or yet-to-be-discovered high-penetrance genes and environmental risk factors. A family history of PC, a high number of nevi and melanomas predict a monogenic predisposition to melanoma.
Collapse
Affiliation(s)
- Giada Ferrara
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Verona, Italy
| | - Salvatore Paiella
- Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Giulio Settanni
- Pathology Unit, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, Verona, Italy
| | - Melissa Frizziero
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, Manchester, UK
| | - Paolo Rosina
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Verona, Italy
| | - Valeria Viassolo
- Medical Genetics, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, Verona, Italy
| |
Collapse
|
5
|
Holic L. Common skin cancers and their association with other non-cutaneous primary malignancies: a review of the literature. Med Oncol 2024; 41:157. [PMID: 38758457 DOI: 10.1007/s12032-024-02385-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/16/2024] [Indexed: 05/18/2024]
Abstract
It has long been recognized that a history of skin cancer puts one at risk for additional primary skin cancers. However, more variable data exists for the risk of developing a non-cutaneous primary cancer following a diagnosis of skin cancer. The data are most variable for Basal Cell Carcinoma (BCC), the most common and least aggressive type of skin cancer. While early studies imply that BCC does not impart a larger risk of other primary non-cutaneous cancers, more recent studies with larger populations suggest otherwise. The cancers most significantly associated with BCC are lip, oropharyngeal, and salivary gland cancer. There is also burgeoning evidence to suggest a link between BCC and prostate, breast, and colorectal cancer, but more data are needed to draw a concrete conclusion. Squamous Cell Carcinoma (SCC), the second most common type of skin cancer, has a slightly more defined risk to other non-cutaneous primary malignancies. There is a notable link between SCC and non-Hodgkin's lymphoma (NHL), possibly due to immunosuppression. There is also an increased risk of other cancers derived from squamous epithelium following SCC, including oropharyngeal, lip, and salivary gland cancer. Some studies also suggest an increased risk of respiratory tract cancer following SCC, possibly due to shared risk factors. Melanoma, a more severe type of skin cancer, shows a well-defined risk of additional primary non-cutaneous malignancies. The most significant of these risks include NHL, thyroid cancer, prostate cancer, and breast cancer along with a host of other cancers. Each of these three main skin cancer types has a profile of genetic mutations that have also been linked to non-cutaneous malignancies. In this review, we discuss a selection of these genes to highlight the complex interplay between different tumorigenesis processes.
Collapse
Affiliation(s)
- Lindsay Holic
- Chicago Medical School at Rosalind Franklin University, North Chicago, IL, USA.
| |
Collapse
|
6
|
Narod SA, Metcalfe K, Finch A, Chan AW, Armel SR, Aeilts A, Eisen A, Karlan B, Bordeleau L, Tung N, Foulkes WD, Neuhausen SL, Eng C, Olopade O, Zakalik D, Couch F, Cullinane C, Pal T, Sun P, Kotsopoulos J. The risk of skin cancer in women who carry BRCA1 or BRCA2 mutations. Hered Cancer Clin Pract 2024; 22:7. [PMID: 38741145 DOI: 10.1186/s13053-024-00277-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/02/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND It has not been clearly established if skin cancer or melanoma are manifestations of BRCA1 or BRCA2 mutation carrier status. Estimating the risk of skin cancer is an important step towards developing screening recommendations. METHODS We report the findings of a prospective cohort study of 6,207 women from North America who carry BRCA1 or BRCA2 mutations. Women were followed from the date of baseline questionnaire to the diagnosis of skin cancer, to age 80 years, death from any cause, or the date of last follow-up. RESULTS During the mean follow-up period of eight years, 3.7% of women with a BRCA1 mutation (133 of 3,623) and 3.8% of women with a BRCA2 mutation (99 of 2,584) reported a diagnosis of skin cancer (including both keratinocyte carcinomas and melanoma). The cumulative risk of all types of skin cancer from age 20 to 80 years was 14.1% for BRCA1 carriers and 10.7% for BRCA2 carriers. The cumulative risk of melanoma was 2.5% for BRCA1 carriers and 2.3% for BRCA2 carriers, compared to 1.5% for women in the general population in the United States. The strongest risk factor for skin cancer was a prior diagnosis of skin cancer. CONCLUSION The risk of non-melanoma skin cancer in women who carry a mutation in BRCA1 or BRCA2 is similar to that of non-carrier women. The risk of melanoma appears to be slightly elevated. We suggest that a referral to a dermatologist or primary care provider for BRCA mutation carriers for annual skin examination and counselling regarding limiting UV exposure, the use of sunscreen and recognizing the early signs of melanoma might be warranted, but further studies are necessary.
Collapse
Affiliation(s)
- Steven A Narod
- Women's College Research Institute, Women's College Hospital, 76 Grenville St, M5S 1B1, Toronto, ON, Canada.
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
| | - Kelly Metcalfe
- Women's College Research Institute, Women's College Hospital, 76 Grenville St, M5S 1B1, Toronto, ON, Canada
- Bloomberg School of Nursing, University of Toronto, Toronto, ON, Canada
| | - Amy Finch
- Women's College Research Institute, Women's College Hospital, 76 Grenville St, M5S 1B1, Toronto, ON, Canada
| | - An-Wen Chan
- Women's College Research Institute, Women's College Hospital, 76 Grenville St, M5S 1B1, Toronto, ON, Canada
- Division of Dermatology, Department of Medicine, University of Toronto, Toronto, Canada
| | - Susan Randall Armel
- Princess Margaret Hospital, Familial Cancer Clinic, University Health Network, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Amber Aeilts
- Division of Human Genetics, Comprehensive Cancer Center, the Ohio State University Medical Center, Columbus, OH, USA
| | | | - Beth Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Louise Bordeleau
- Department of Oncology, McMaster University, Hamilton, ON, Canada
| | - Nadine Tung
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - William D Foulkes
- Program in Cancer Genetics, Department of Oncology and Human Genetics, McGill University, Montréal, QC, Canada
| | - Susan L Neuhausen
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Charis Eng
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Olufunmilayo Olopade
- Department of Medicine and Human Genetics, University of Chicago, Chicago, IL, USA
| | - Dana Zakalik
- Cancer Genetics Program, Beaumont Hospital, Royal Oak, MI, USA
| | - Fergus Couch
- Division of Experimental Pathology and Laboratory Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Tuya Pal
- Division of Genetics, Department of Medicine, Vanderbilt University Medical Centre and Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Ping Sun
- Women's College Research Institute, Women's College Hospital, 76 Grenville St, M5S 1B1, Toronto, ON, Canada
| | - Joanne Kotsopoulos
- Women's College Research Institute, Women's College Hospital, 76 Grenville St, M5S 1B1, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
7
|
Stefanoudakis D, Frountzas M, Schizas D, Michalopoulos NV, Drakaki A, Toutouzas KG. Significance of TP53, CDKN2A, SMAD4 and KRAS in Pancreatic Cancer. Curr Issues Mol Biol 2024; 46:2827-2844. [PMID: 38666907 PMCID: PMC11049225 DOI: 10.3390/cimb46040177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/16/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
The present review demonstrates the major tumor suppressor genes, including TP53, CDKN2A and SMAD4, associated with pancreatic cancer. Each gene's role, prevalence and impact on tumor development and progression are analyzed, focusing on the intricate molecular landscape of pancreatic cancer. In addition, this review underscores the prognostic significance of specific mutations, such as loss of TP53, and explores some potential targeted therapies tailored to these molecular signatures. The findings highlight the importance of genomic analyses for risk assessment, early detection and the design of personalized treatment approaches in pancreatic cancer. Overall, this review provides a comprehensive analysis of the molecular intricacies of pancreatic tumors, paving the way for more effective and tailored therapeutic interventions.
Collapse
Affiliation(s)
- Dimitrios Stefanoudakis
- First Propaedeutic Department of Surgery, Hippocration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (N.V.M.)
| | - Maximos Frountzas
- First Propaedeutic Department of Surgery, Hippocration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (N.V.M.)
| | - Dimitrios Schizas
- First Department of Surgery, Laikon General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Nikolaos V. Michalopoulos
- First Propaedeutic Department of Surgery, Hippocration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (N.V.M.)
| | - Alexandra Drakaki
- Division of Hematology and Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Konstantinos G. Toutouzas
- First Propaedeutic Department of Surgery, Hippocration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (N.V.M.)
| |
Collapse
|
8
|
Wunderlich K, Suppa M, Gandini S, Lipski J, White JM, Del Marmol V. Risk Factors and Innovations in Risk Assessment for Melanoma, Basal Cell Carcinoma, and Squamous Cell Carcinoma. Cancers (Basel) 2024; 16:1016. [PMID: 38473375 DOI: 10.3390/cancers16051016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/22/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Skin cancer is the most frequently diagnosed cancer globally and is preventable. Various risk factors contribute to different types of skin cancer, including melanoma, basal cell carcinoma, and squamous cell carcinoma. These risk factors encompass both extrinsic, such as UV exposure and behavioral components, and intrinsic factors, especially involving genetic predisposition. However, the specific risk factors vary among the skin cancer types, highlighting the importance of precise knowledge to facilitate appropriate early diagnosis and treatment for at-risk individuals. Better understanding of the individual risk factors has led to the development of risk scores, allowing the identification of individuals at particularly high risk. These advances contribute to improved prevention strategies, emphasizing the commitment to mitigating the impact of skin cancer.
Collapse
Affiliation(s)
- K Wunderlich
- Department of Dermatology, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - M Suppa
- Department of Dermatology, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
- Department of Dermatology, Institute Jules Bordet, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - S Gandini
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, European Institute of Oncology, IRCCS, 20139 Milan, Italy
| | - J Lipski
- Department of Dermatology, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - J M White
- Department of Dermatology, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - V Del Marmol
- Department of Dermatology, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
- Department of Dermatology, Institute Jules Bordet, Université Libre de Bruxelles, 1070 Brussels, Belgium
| |
Collapse
|
9
|
Shao L, Zhao Y, Heinrich M, Prieto-Garcia JM, Manzoni C. Active natural compounds perturb the melanoma risk-gene network. G3 (BETHESDA, MD.) 2024; 14:jkad274. [PMID: 38035793 PMCID: PMC10849364 DOI: 10.1093/g3journal/jkad274] [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: 10/03/2023] [Revised: 10/27/2023] [Accepted: 11/09/2023] [Indexed: 12/02/2023]
Abstract
Cutaneous melanoma is an aggressive type of skin cancer with a complex genetic landscape caused by the malignant transformation of melanocytes. This study aimed at providing an in silico network model based on the systematic profiling of the melanoma-associated genes considering germline mutations, somatic mutations, and genome-wide association study signals accounting for a total of 232 unique melanoma risk genes. A protein-protein interaction network was constructed using the melanoma risk genes as seeds and evaluated to describe the functional landscape in which the melanoma genes operate within the cellular milieu. Not only were the majority of the melanoma risk genes able to interact with each other at the protein level within the core of the network, but this showed significant enrichment for genes whose expression is altered in human melanoma specimens. Functional annotation showed the melanoma risk network to be significantly associated with processes related to DNA metabolism and telomeres, DNA damage and repair, cellular ageing, and response to radiation. We further explored whether the melanoma risk network could be used as an in silico tool to predict the efficacy of anti-melanoma phytochemicals, that are considered active molecules with potentially less systemic toxicity than classical cytotoxic drugs. A significant portion of the melanoma risk network showed differential expression when SK-MEL-28 human melanoma cells were exposed to the phytochemicals harmine and berberine chloride. This reinforced our hypothesis that the network modeling approach not only provides an alternative way to identify molecular pathways relevant to disease but it may also represent an alternative screening approach to prioritize potentially active compounds.
Collapse
Affiliation(s)
- Luying Shao
- Department of Pharmaceutical and Biological Chemistry, UCL School of Pharmacy, WC1N 1AX London, UK
| | - Yibo Zhao
- Department of Pharmacology, UCL School of Pharmacy, WC1N 1AX London, UK
| | - Michael Heinrich
- Department of Pharmaceutical and Biological Chemistry, UCL School of Pharmacy, WC1N 1AX London, UK
- Chinese Medicine Research Center, and Department of Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung City 404333, Taiwan
| | - Jose M Prieto-Garcia
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, L3 3AF Liverpool, UK
| | - Claudia Manzoni
- Department of Pharmacology, UCL School of Pharmacy, WC1N 1AX London, UK
| |
Collapse
|
10
|
Atkinson C, McInerney-Leo AM, Proctor M, Lanagan C, Stevenson AJ, Dehkhoda F, Caole M, Maas E, Ainger S, Pritchard AL, Johansson PA, Leo P, Hayward NK, Sturm RA, Duncan EL, Gabrielli B. The ATM Ser49Cys Variant Effects ATM Function as a Regulator of Oncogene-Induced Senescence. Int J Mol Sci 2024; 25:1664. [PMID: 38338943 PMCID: PMC10855307 DOI: 10.3390/ijms25031664] [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: 11/28/2023] [Revised: 01/15/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
An apical component of the cell cycle checkpoint and DNA damage repair response is the ataxia-telangiectasia mutated (ATM) Ser/Thr protein kinase. A variant of ATM, Ser49Cys (rs1800054; minor allele frequency = 0.011), has been associated with an elevated risk of melanoma development; however, the functional consequence of this variant is not defined. ATM-dependent signalling in response to DNA damage has been assessed in a panel of patient-derived lymphoblastoid lines and primary human melanocytic cell strains heterozygous for the ATM Ser49Cys variant allele. The ATM Ser49Cys allele appears functional for acute p53-dependent signalling in response to DNA damage. Expression of the variant allele did reduce the efficacy of oncogene expression in inducing senescence. These findings demonstrate that the ATM 146C>G Ser49Cys allele has little discernible effect on the acute response to DNA damage but has reduced function observed in the chronic response to oncogene over-expression. Analysis of melanoma, naevus and skin colour genomics and GWAS analyses have demonstrated no association of this variant with any of these outcomes. The modest loss of function detected suggest that the variant may act as a modifier of other variants of ATM/p53-dependent signalling.
Collapse
Affiliation(s)
- Caroline Atkinson
- Mater Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Aideen M. McInerney-Leo
- Dermatology Research Centre, Frazer Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Martina Proctor
- Mater Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Catherine Lanagan
- Mater Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | | | - Farhad Dehkhoda
- Dermatology Research Centre, Frazer Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Mary Caole
- Mater Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Ellie Maas
- Dermatology Research Centre, Frazer Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Stephen Ainger
- Dermatology Research Centre, Frazer Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Antonia L. Pritchard
- Queensland Institute for Medical Research Berghofer, Brisbane, QLD 4006, Australia
| | - Peter A. Johansson
- Queensland Institute for Medical Research Berghofer, Brisbane, QLD 4006, Australia
| | - Paul Leo
- Centre of Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Nicholas K. Hayward
- Queensland Institute for Medical Research Berghofer, Brisbane, QLD 4006, Australia
| | - Richard A. Sturm
- Dermatology Research Centre, Frazer Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Emma L. Duncan
- Department of Twin Research and Genetic Epidemiology, School of Life Course & Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, UK
| | - Brian Gabrielli
- Mater Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| |
Collapse
|
11
|
Sortino AMF, Soares de Sá BC, Martins MA, Bertolli E, de Paula RB, Lopes Pinto CA, David Filho WJ, Tavoloni Braga JC, Duprat Neto JP, Carraro DM, Curado MP. Multiple Primary Melanoma: A Five-Year Prospective Single-Center Follow-Up Study of Two MC1R R/R Genotype Carriers. Life (Basel) 2023; 13:2102. [PMID: 37895483 PMCID: PMC10608495 DOI: 10.3390/life13102102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/10/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Multiple primary melanoma (MPM) is a diagnostic challenge even with ancillary imaging technologies available to dermatologists. In selected patients' phenotypes, the use of imaging approaches can help better understand lesion characteristics, and aid in early diagnosis and management. METHODS Under a 5-year prospective single-center follow-up, 58 s primary melanomas (SPMs) were diagnosed in two first-degree relatives, with fair skin color, red hair, green eyes, and personal history of one previous melanoma each. Patients' behavior and descriptive demographic data were collected from medical records. The information on the first two primary melanomas (PMs) were retrieved from pathology reports. The characteristics of 60 melanomas were collected from medical records, video dermoscopy software, and pathology reports. Reflectance confocal microscopy (RCM) was performed prior to excision of 22 randomly selected melanomas. RESULTS From February 2018 to May 2023, two patients underwent a pooled total of 214 excisional biopsies of suspect lesions, resulting in a combined benign versus malignant treatment ratio (NNT) of 2.0:1.0. The number of moles excised for each melanoma diagnosed (NNE) was 1.7:1.0 and 6.9:1.0 for the female and male patient respectively. The in-situ melanoma/invasive melanoma ratio (IIR) demonstrated a higher proportion of in-situ melanomas for both patients. From June 2018 to May 2023, a total of 58 SPMs were detected by the combination of total body skin exam (TBSE), total body skin photography (TBSP), digital dermoscopy (DD), and sequential digital dermoscopy imaging (SDDI) via comparative approach. The younger patient had her PM one month prior to the second and third cutaneous melanomas (CMs), characterizing a case of synchronous primary CM. The male older relative had a total of 7 nonsynchronous melanomas. CONCLUSIONS This CM cohort is composed of 83.3% in-situ melanoma and 16.7% invasive melanoma. Both patients had a higher percentage of SPM with clinical nevus-like morphology (84.5%), global dermoscopic pattern of asymmetric multiple component (60.3%) and located on the lower limbs (46.6%). When RCM was performed prior to excision, 81% of SPM had features suggestive of malignancy. As well, invasive melanomas were more frequent in the lower limbs (40%). In the multivariate model, for the two high-risk patients studied, the chance of a not associated with nevus ("de novo") invasive SPM diagnosis is 25 times greater than the chance of a diagnosis of a nevus-associated invasive SPM.
Collapse
Affiliation(s)
- Ana Maria Fagundes Sortino
- Clínica Dermatológica Dermatis, Rua Joaquim Floriano 466, Itaim Bibi, São Paulo 04534-002, SP, Brazil
- Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, Bela Vista, São Paulo 01308-050, SP, Brazil
| | | | - Marcos Alberto Martins
- Centro Universitário Saúde ABC, Surgery Department, Avenida Lauro Gomes 2000, Vila Sacadura Cabral, Santo André 09060-870, SP, Brazil
| | - Eduardo Bertolli
- Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, Bela Vista, São Paulo 01308-050, SP, Brazil
- A Beneficência Portuguesa de São Paulo–BP Mirante, Rua Martiniano de Carvalho 965, Bela Vista, São Paulo 01323-001, SP, Brazil
| | - Rafaela Brito de Paula
- AC Camargo Cancer Center, Rua Pires da Mota 1.167, Aclimação, São Paulo 01529-001, SP, Brazil
| | | | - Waldec Jorge David Filho
- Hospital Alemão Oswaldo-Cruz, Rua Treze de Maio 1815, Bela Vista, São Paulo 01323-903, SP, Brazil;
| | | | | | - Dirce Maria Carraro
- AC Camargo Cancer Center, Rua Pires da Mota 1.167, Aclimação, São Paulo 01529-001, SP, Brazil
| | - Maria Paula Curado
- AC Camargo Cancer Center, Rua Pires da Mota 1.167, Aclimação, São Paulo 01529-001, SP, Brazil
| |
Collapse
|
12
|
Sargen MR, Kim J, Potjer TP, Velthuizen ME, Martir-Negron AE, Odia Y, Helgadottir H, Hatton JN, Haley JS, Thone G, Widemann BC, Gross AM, Yohe ME, Kaplan RN, Shern JF, Sundby RT, Astiazaran-Symonds E, Yang XR, Carey DJ, Tucker MA, Stewart DR, Goldstein AM. Estimated Prevalence, Tumor Spectrum, and Neurofibromatosis Type 1-Like Phenotype of CDKN2A-Related Melanoma-Astrocytoma Syndrome. JAMA Dermatol 2023; 159:1112-1118. [PMID: 37585199 PMCID: PMC10433137 DOI: 10.1001/jamadermatol.2023.2621] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/14/2023] [Indexed: 08/17/2023]
Abstract
Importance Knowledge about the prevalence and tumor types of CDKN2A-related melanoma-astrocytoma syndrome (MAS) is limited and could improve disease recognition. Objective To estimate the prevalence and describe the tumor types of MAS. Design, Setting, and Participants This retrospective cohort study analyzed all available MAS cases from medical centers in the US (2 sites) and Europe (2 sites) and from biomedical population genomic databases (UK Biobank [United Kingdom], Geisinger MyCode [US]) between January 1, 1976, and December 31, 2020. Patients with MAS with CDKN2A germline pathogenic variants and 1 or more neural tumors were included. Data were analyzed from June 1, 2022, to January 31, 2023. Main Outcomes and Measures Disease prevalence and tumor frequency. Results Prevalence of MAS ranged from 1 in 170 503 (n = 1 case; 95% CI, 1:30 098-1:965 887) in Geisinger MyCode (n = 170 503; mean [SD] age, 58.9 [19.1] years; 60.6% women; 96.2% White) to 1 in 39 149 (n = 12 cases; 95% CI, 1:22 396-1:68 434) in UK Biobank (n = 469 789; mean [SD] age, 70.0 [8.0] years; 54.2% women; 94.8% White). Among UK Biobank patients with MAS (n = 12) identified using an unbiased genomic ascertainment approach, brain neoplasms (4 of 12, 33%; 1 glioblastoma, 1 gliosarcoma, 1 astrocytoma, 1 unspecified type) and schwannomas (3 of 12, 25%) were the most common malignant and benign neural tumors, while cutaneous melanoma (2 of 12, 17%) and head and neck squamous cell carcinoma (2 of 12, 17%) were the most common nonneural malignant neoplasms. In a separate case series of 14 patients with MAS from the US and Europe, brain neoplasms (4 of 14, 29%; 2 glioblastomas, 2 unspecified type) and malignant peripheral nerve sheath tumor (2 of 14, 14%) were the most common neural cancers, while cutaneous melanoma (4 of 14, 29%) and sarcomas (2 of 14, 14%; 1 liposarcoma, 1 unspecified type) were the most common nonneural cancers. Cutaneous neurofibromas (7 of 14, 50%) and schwannomas (2 of 14, 14%) were also common. In 1 US family, a father and son with MAS had clinical diagnoses of neurofibromatosis type 1 (NF1). Genetic testing of the son detected a pathogenic CDKN2A splicing variant (c.151-1G>C) and was negative for NF1 genetic alterations. In UK Biobank, 2 in 150 (1.3%) individuals with clinical NF1 diagnoses had likely pathogenic variants in CDKN2A, including 1 individual with no detected variants in the NF1 gene. Conclusions and Relevance This cohort study estimates the prevalence and describes the tumors of MAS. Additional studies are needed in genetically diverse populations to further define population prevalence and disease phenotypes.
Collapse
Affiliation(s)
- Michael R. Sargen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Jung Kim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Thomas P. Potjer
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Mary E. Velthuizen
- Division Laboratories, Pharmacy and Biomedical Genetics, Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Yazmin Odia
- Miami Cancer Institute, Baptist Health South Florida, Miami
| | - Hildur Helgadottir
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Jessica N. Hatton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Jeremy S. Haley
- Department of Genomic Health, Geisinger Clinic, Geisinger Health System, Danville, Pennsylvania
| | - Gretchen Thone
- Department of Genomic Health, Geisinger Clinic, Geisinger Health System, Danville, Pennsylvania
| | - Brigitte C. Widemann
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Andrea M. Gross
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Marielle E. Yohe
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, Frederick, Maryland
| | - Rosandra N. Kaplan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jack F. Shern
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - R. Taylor Sundby
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Xiaohong R. Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - David J. Carey
- Department of Genomic Health, Geisinger Clinic, Geisinger Health System, Danville, Pennsylvania
| | - Margaret A. Tucker
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Douglas R. Stewart
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Alisa M. Goldstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| |
Collapse
|
13
|
Izol Özmen H. Publication Trends and Hot Topics in Dysplastic Nevus Research: A 30-Year Bibliometric Analysis. Dermatol Pract Concept 2023; 13:dpc.1304a266. [PMID: 37992349 PMCID: PMC10656159 DOI: 10.5826/dpc.1304a266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2023] [Indexed: 11/24/2023] Open
Abstract
INTRODUCTION Dysplastic nevi are pigmented lesions that exhibit clinical and histological features of both common nevi and melanoma. In recent years, there has been an increase in publications on dysplastic nevi. Bibliometric analysis is a method of evaluating trends in large number of publications and identifying popular topics. OBJECTIVES The objective of this study is to provide an overview of the landscape of publications related to dysplastic nevi, visualize trends and identify popular topics in the literature. METHODS Thomson Reuters' Web of Science database was searched with the following query in title, abstract or keywords: TS = ("dysplastic nevus" OR "clark nevus" OR "atypical nevus" OR "dysplastic nevi" OR "clark nevi" OR "atypical nevi"). Time span was set to 1992-2022. Document type was set to Article. Titles, authors, abstracts, institutions, countries, journals, references, and the citation information were recorded. RESULTS Although the number of publications has declined over time, the USA remains the leading contributor to published articles. Key clusters of frequently used keywords were identified. The Journal of the American Academy of Dermatology had the highest number of published titles. Country and journal analysis were supplemented by co-citation and co-cited reference cluster analysis. Burst analyses revealed authors like Kittler, Argenziano, and Gandini as significant contributors, with their works receiving strong citation bursts extending until the end of the study period. CONCLUSIONS This bibliometric analysis revealed trends and interest pockets in the literature pertaining to dysplastic nevi and melanoma. This study aids in understanding the current research landscape and highlights potential future directions in this field.
Collapse
Affiliation(s)
- Hazal Izol Özmen
- Department of Pathology, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| |
Collapse
|
14
|
Khan SU, Ullah Z, Shaukat H, Unab S, Jannat S, Ali W, Ali A, Irfan M, Khan MF, Cervantes-Villagrana RD. TP53 and its Regulatory Genes as Prognosis of Cutaneous Melanoma. Cancer Inform 2023; 22:11769351231177267. [PMID: 37667731 PMCID: PMC10475268 DOI: 10.1177/11769351231177267] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 09/06/2023] Open
Abstract
The present study was the first comprehensive investigation of genetic mutation and expression levels of the p53 signaling genes in cutaneous melanoma through various genetic databases providing large datasets. The mutational landscape of p53 and its signaling genes was higher than expected, with TP53 followed by CDKN2A being the most mutated gene in cutaneous melanoma. Furthermore, the expression analysis showed that TP53, MDM2, CDKN2A, and TP53BP1 were overexpressed, while MDM4 and CDKN2B were under-expressed in cutaneous melanoma. Overall, TCGA data revealed that among all the other p53 signaling proteins, CDKN2A was significantly higher in both sun and non-sun-exposed healthy tissues than in melanoma. Likewise, MDM4 and TP53BP1 expressions were markedly greater in non-sun-exposed healthy tissues compared to other groups. However, CDKN2B expression was higher in the sun-exposed healthy tissues than in other tissues. In addition, various genes were expressed significantly differently among males and females. In addition, CDKN2A was highly expressed in the SK-MEL-30 skin cancer cell line, whereas, Immune cell type expression analysis revealed that the MDM4 was highly expressed in naïve B-cells. Furthermore, all six genes were significantly overexpressed in extraordinarily overweight or obese tumor tissues compared to healthy tissues. MDM2 expression and tumor stage were closely related. There were differences in gene expression across patient age groups and positive nodal status. TP53 showed a positive correlation with B cells, MDM2 with CD8+T cells, macrophages and neutrophils, and MDM4 with neutrophils. CDKN2A/B had a non-significant correlation with all six types of immune cells. However, TP53BP1 was positively correlated with all five types of immune cells except B cells. Only TP53, MDM2, and CDKN2A had a role in cutaneous melanoma-specific tumor immunity. All TP53 and its regulating genes may be predictive for prognosis. The results of the present study need to be validated through future screening, in vivo, and in vitro studies.
Collapse
Affiliation(s)
- Safir Ullah Khan
- Department of Cell Biology, Center for Research and Advanced Studies, National Polytechnic Institute, Mexico City, Mexico
| | - Zahid Ullah
- Department of Software Engineering, Abasyn University Peshawar, Peshawar, Pakistan
| | - Hadia Shaukat
- Department of Zoology, Women University of Swabi, Swabi, Khyber Pakhtunkhwa, Pakistan
| | - Sheeza Unab
- Department of Zoology, University of Mianwali, Mianwali, Pakistan
| | - Saba Jannat
- Department of Zoology, Women University of Swabi, Swabi, Khyber Pakhtunkhwa, Pakistan
| | - Waqar Ali
- Biochemical and Biotechnological Sciences Università degli studi della Campania Luigi Vanvitelli, Caserta, Campania, Italy
| | - Amir Ali
- Nanosciences and Nanotechnology Program, Center for Research and Advanced Studies, National Polytechnic Institute, Mexico City, Mexico
| | - Muhammad Irfan
- Department of Zoology, Wildlife and Fisheries, Pir Mehr Ali Shah Arid Agriculture University Rawalpindi, Punjab, Pakistan
| | | | | |
Collapse
|
15
|
Cakir A, Elcin G, Kilickap S, Gököz Ö, Taskiran ZE, Celik İ. Phenotypic and Genetic Features that Differ Between Hereditary and Sporadic Melanoma: Results of a Preliminary Study from a Single Center from Turkey. Dermatol Pract Concept 2023; 13:e2023146. [PMID: 37557112 PMCID: PMC10412028 DOI: 10.5826/dpc.1303a146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2023] [Indexed: 08/11/2023] Open
Abstract
INTRODUCTION Most melanoma patients under our supervision lack characteristic phenotypic features for melanoma. In contrast, history of cancers other than melanoma and early age at onset were common. This observation was in favor of hereditary melanoma. OBJECTIVES To search for the phenotypic and genetic features that differ between sporadic and hereditary melanomas. METHODS In order to reveal phenotypic features, detailed physical exam was conducted to all melanoma patients (N = 43) and for genetic features. CDKN2A and MC1R mutations were detected with Sanger sequencing method. Assignment to hereditary and sporadic groups was done according to the "melanoma cancer syndrome assessment tool". Patients who were diagnosed before the age of 50 were also assigned to the hereditary melanoma group. RESULTS Thirty-one patients were assigned to the hereditary group and 12 to the sporadic group. Fair eye color was statistically significantly higher in the sporadic group (P = 0.000). CDKN2A was detected in only 1 patient in the hereditary group. MC1R mutations were found in 12 out of 13 (92.3%) in the hereditary group with a score =3 points, 13 out of 18 (72.2%) in the early age at onset group and 5 out of 12 (41.7%) in the sporadic group (P = 0.024). CONCLUSIONS Incidence of CDKN2A mutations in our hereditary group is in accordance with the reported incidences from Mediterranean countries. The difference between the hereditary and sporadic groups in terms of MC1R mutations supports the idea that MC1R genetic testing might help to determine patients with higher risk for hereditary melanoma.
Collapse
Affiliation(s)
- Aysel Cakir
- Hacettepe University Faculty of Medicine, Department of Dermatology and Venerology, Ankara, Turkey
| | - Gonca Elcin
- Hacettepe University Faculty of Medicine, Department of Dermatology and Venerology, Ankara, Turkey
| | - Saadettin Kilickap
- Hacettepe University Faculty of Medicine, Department of Medical Oncology, Ankara, Turkey
| | - Özay Gököz
- Hacettepe University Faculty of Medicine, Department of Medical Pathology, Ankara, Turkey
| | - Zihni Ekim Taskiran
- Hacettepe University Faculty of Medicine, Department of Medical Genetics, Ankara, Turkey
| | - İsmail Celik
- Hacettepe University Faculty of Medicine, Department of Medical Oncology, Ankara, Turkey
| |
Collapse
|
16
|
Rahimi A, Esmaeili Y, Dana N, Dabiri A, Rahimmanesh I, Jandaghain S, Vaseghi G, Shariati L, Zarrabi A, Javanmard SH, Cordani M. A comprehensive review on novel targeted therapy methods and nanotechnology-based gene delivery systems in melanoma. Eur J Pharm Sci 2023:106476. [PMID: 37236377 DOI: 10.1016/j.ejps.2023.106476] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023]
Abstract
Melanoma, a malignant form of skin cancer, has been swiftly increasing in recent years. Although there have been significant advancements in clinical treatment underlying a well-understanding of melanoma-susceptible genes and the molecular basis of melanoma pathogenesis, the permanency of response to therapy is frequently constrained by the emergence of acquired resistance and systemic toxicity. Conventional therapies, including surgical resection, chemotherapy, radiotherapy, and immunotherapy, have already been used to treat melanoma and are dependent on the cancer stage. Nevertheless, ineffective side effects and the heterogeneity of tumors pose major obstacles to the therapeutic treatment of malignant melanoma through such strategies. In light of this, advanced therapies including nucleic acid therapies (ncRNA, aptamers), suicide gene therapies, and gene therapy using tumor suppressor genes, have lately gained immense attention in the field of cancer treatment. Furthermore, nanomedicine and targeted therapy based on gene editing tools have been applied to the treatment of melanoma as potential cancer treatment approaches nowadays. Indeed, nanovectors enable delivery of the therapeutic agents into the tumor sites by passive or active targeting, improving therapeutic efficiency and minimizing adverse effects. Accordingly, in this review, we summarized the recent findings related to novel targeted therapy methods as well as nanotechnology-based gene systems in melanoma. We also discussed current issues along with potential directions for future research, paving the way for the next-generation of melanoma treatments.
Collapse
Affiliation(s)
- Azadeh Rahimi
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yasaman Esmaeili
- Biosensor Research Center, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Nasim Dana
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arezou Dabiri
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Setareh Jandaghain
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Golnaz Vaseghi
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran; Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8158388994, Iran
| | - Laleh Shariati
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering & Natural Sciences, Istinye University, Istanbul 34396, Turkey
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University, 28040 Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain.
| |
Collapse
|
17
|
Oh KS, Mahalingam M. Melanoma and Glioblastoma-Not a Serendipitous Association. Adv Anat Pathol 2023; 30:00125480-990000000-00051. [PMID: 36624550 DOI: 10.1097/pap.0000000000000393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Recently, we came across a patient with malignant melanoma and primary glioblastoma. Given this, we parsed the literature to ascertain the relationship, if any, between these 2 malignancies. We begin with a brief overview of melanoma and glioma in isolation followed by a chronologic overview of case reports and epidemiologic studies documenting both neoplasms. This is followed by studies detailing genetic abnormalities common to both malignancies with a view to identifying unifying genetic targets for therapeutic strategies as well as to explore the possibility of a putative association and an inherited cancer susceptibility trait. From a scientific perspective, we believe we have provided evidence favoring an association between melanoma and glioma. Future studies that include documentation of additional cases, as well as a detailed molecular analyses, will lend credence to our hypothesis that the co-occurrence of these 2 conditions is likely not serendipitous.
Collapse
Affiliation(s)
- Kei Shing Oh
- Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL
| | - Meera Mahalingam
- Dermatopathology Section, Department of Pathology and Laboratory Medicine, VA-Integrated-Service-Network-1 (VISN1), West Roxbury, MA
| |
Collapse
|
18
|
Newell F, Johansson PA, Wilmott JS, Nones K, Lakis V, Pritchard AL, Lo SN, Rawson RV, Kazakoff SH, Colebatch AJ, Koufariotis LT, Ferguson PM, Wood S, Leonard C, Law MH, Brooks KM, Broit N, Palmer JM, Couts KL, Vergara IA, Long GV, Barbour AP, Nieweg OE, Shivalingam B, Robinson WA, Stretch JR, Spillane AJ, Saw RP, Shannon KF, Thompson JF, Mann GJ, Pearson JV, Scolyer RA, Waddell N, Hayward NK. Comparative Genomics Provides Etiologic and Biological Insight into Melanoma Subtypes. Cancer Discov 2022; 12:2856-2879. [PMID: 36098958 PMCID: PMC9716259 DOI: 10.1158/2159-8290.cd-22-0603] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/01/2022] [Accepted: 09/02/2022] [Indexed: 01/12/2023]
Abstract
Melanoma is a cancer of melanocytes, with multiple subtypes based on body site location. Cutaneous melanoma is associated with skin exposed to ultraviolet radiation; uveal melanoma occurs in the eyes; mucosal melanoma occurs in internal mucous membranes; and acral melanoma occurs on the palms, soles, and nail beds. Here, we present the largest whole-genome sequencing study of melanoma to date, with 570 tumors profiled, as well as methylation and RNA sequencing for subsets of tumors. Uveal melanoma is genomically distinct from other melanoma subtypes, harboring the lowest tumor mutation burden and with significantly mutated genes in the G-protein signaling pathway. Most cutaneous, acral, and mucosal melanomas share alterations in components of the MAPK, PI3K, p53, p16, and telomere pathways. However, the mechanism by which these pathways are activated or inactivated varies between melanoma subtypes. Additionally, we identify potential novel germline predisposition genes for some of the less common melanoma subtypes. SIGNIFICANCE This is the largest whole-genome analysis of melanoma to date, comprehensively comparing the genomics of the four major melanoma subtypes. This study highlights both similarities and differences between the subtypes, providing insights into the etiology and biology of melanoma. This article is highlighted in the In This Issue feature, p. 2711.
Collapse
Affiliation(s)
- Felicity Newell
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Corresponding Authors: Felicity Newell, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia. Phone: 61-7-3845-3965; E-mail: ; Richard A. Scolyer, Melanoma Institute Australia, 40 Rockland Road, Wollstonecraft, Sydney, NSW 2065, Australia. Phone: 61-2-9515-7011; E-mail: ; and Nicola Waddell, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia. Phone: 61-7-3845-3538;
| | - Peter A. Johansson
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - James S. Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Katia Nones
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Vanessa Lakis
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Antonia L. Pritchard
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Department of Genetics and Immunology, Division of Biomedical Science, University of the Highlands and Islands, Inverness, Scotland, United Kingdom
| | - Serigne N. Lo
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Robert V. Rawson
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, New South Wales, Australia
| | | | - Andrew J. Colebatch
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, New South Wales, Australia
| | | | - Peter M. Ferguson
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, New South Wales, Australia
| | - Scott Wood
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Conrad Leonard
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Matthew H. Law
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kelly M. Brooks
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Natasa Broit
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Q-Gen Cell Therapeutics, Brisbane, Queensland, Australia
| | - Jane M. Palmer
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kasey L. Couts
- Center for Rare Melanomas, University of Colorado Cancer Center, Aurora, Colorado
| | - Ismael A. Vergara
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Georgina V. Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Mater Hospital, North Sydney, New South Wales, Australia.,Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Andrew P. Barbour
- Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Omgo E. Nieweg
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Brindha Shivalingam
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Mater Hospital, North Sydney, New South Wales, Australia.,Department of Neurosurgery, Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia.,Department of Neurosurgery, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - William A. Robinson
- Center for Rare Melanomas, University of Colorado Cancer Center, Aurora, Colorado
| | - Jonathan R. Stretch
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Mater Hospital, North Sydney, New South Wales, Australia.,Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Andrew J. Spillane
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Mater Hospital, North Sydney, New South Wales, Australia.,Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Robyn P.M. Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Mater Hospital, North Sydney, New South Wales, Australia.,Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Kerwin F. Shannon
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - John F. Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Mater Hospital, North Sydney, New South Wales, Australia.,Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Graham J. Mann
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Centre for Cancer Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia.,John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - John V. Pearson
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Richard A. Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, New South Wales, Australia.,Corresponding Authors: Felicity Newell, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia. Phone: 61-7-3845-3965; E-mail: ; Richard A. Scolyer, Melanoma Institute Australia, 40 Rockland Road, Wollstonecraft, Sydney, NSW 2065, Australia. Phone: 61-2-9515-7011; E-mail: ; and Nicola Waddell, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia. Phone: 61-7-3845-3538;
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Corresponding Authors: Felicity Newell, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia. Phone: 61-7-3845-3965; E-mail: ; Richard A. Scolyer, Melanoma Institute Australia, 40 Rockland Road, Wollstonecraft, Sydney, NSW 2065, Australia. Phone: 61-2-9515-7011; E-mail: ; and Nicola Waddell, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia. Phone: 61-7-3845-3538;
| | | |
Collapse
|
19
|
Wong SL, Martiniuc D, Kiuru M. CDKN2A exon 1B deletion predisposing to melanoma and neural system tumour syndrome. Clin Exp Dermatol 2022; 47:2284-2285. [PMID: 35904890 PMCID: PMC9712160 DOI: 10.1111/ced.15354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2022] [Indexed: 11/30/2022]
Abstract
CDKN2A at chromosome positon 9p21 is a tumour suppressor gene encoding the cell cycle regulators p16 and p14ARF. While melanoma is associated with variants affecting both transcripts, families with mutations involving the p14ARF-specific exon 1B may be predisposed to central nervous system tumours. We describe a family with a deletion of exon 1B in CDKN2A, who had multiple cutaneous melanomas, neural tumours and various malignancies.
Collapse
Affiliation(s)
- Samantha L. Wong
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | - Daniela Martiniuc
- Hereditary Cancer Program, Comprehensive Cancer Center, University of California, Davis, Sacramento, CA, USA
| | - Maija Kiuru
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
20
|
Clarysse K, Lacy K. Why, Who and How We Should Screen for Melanoma. CURRENT GENETIC MEDICINE REPORTS 2022. [DOI: 10.1007/s40142-022-00204-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
21
|
Germline Testing for Individuals with Pancreatic Adenocarcinoma and Novel Genetic Risk Factors. Hematol Oncol Clin North Am 2022; 36:943-960. [DOI: 10.1016/j.hoc.2022.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
22
|
Patil P, Pencheva BB, Patil VM, Fangusaro J. Nervous system (NS) Tumors in Cancer Predisposition Syndromes. Neurotherapeutics 2022; 19:1752-1771. [PMID: 36056180 PMCID: PMC9723057 DOI: 10.1007/s13311-022-01277-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2022] [Indexed: 12/13/2022] Open
Abstract
Genetic syndromes which develop one or more nervous system (NS) tumors as one of the manifestations can be grouped under the umbrella term of NS tumor predisposition syndromes. Understanding the underlying pathological pathways at the molecular level has led us to many radical discoveries, in understanding the mechanisms of tumorigenesis, tumor progression, interactions with the tumor microenvironment, and development of targeted therapies. Currently, at least 7-10% of all pediatric cancers are now recognized to occur in the setting of genetic predisposition to cancer or cancer predisposition syndromes. Specifically, the cancer predisposition rate in pediatric patients with NS tumors has been reported to be as high as 15%, though it can approach 50% in certain tumor types (i.e., choroid plexus carcinoma associated with Li Fraumeni Syndrome). Cancer predisposition syndromes are caused by pathogenic variation in genes that primarily function as tumor suppressors and proto-oncogenes. These variants are found in the germline or constitutional DNA. Mosaicism, however, can affect only certain tissues, resulting in varied manifestations. Increased understanding of the genetic underpinnings of cancer predisposition syndromes and the ability of clinical laboratories to offer molecular genetic testing allows for improvement in the identification of these patients. The identification of a cancer predisposition syndrome in a CNS tumor patient allows for changes to medical management to be made, including the initiation of cancer surveillance protocols. Finally, the identification of at-risk biologic relatives becomes feasible through cascade (genetic) testing. These fundamental discoveries have also broadened the horizon of novel therapeutic possibilities and have helped to be better predictors of prognosis and survival. The treatment paradigm of specific NS tumors may also vary based on the patient's cancer predisposition syndrome and may be used to guide therapy (i.e., immune checkpoint inhibitors in constitutional mismatch repair deficiency [CMMRD] predisposition syndrome) [8]. Early diagnosis of these cancer predisposition syndromes is therefore critical, in both unaffected and affected patients. Genetic counselors are uniquely trained master's level healthcare providers with a focus on the identification of hereditary disorders, including hereditary cancer, or cancer predisposition syndromes. Genetic counseling, defined as "the process of helping people understand and adapt to the medical, psychological and familial implications of genetic contributions to disease" plays a vital role in the adaptation to a genetic diagnosis and the overall management of these diseases. Cancer predisposition syndromes that increase risks for NS tumor development in childhood include classic neurocutaneous disorders like neurofibromatosis type 1 and type 2 (NF1, NF2) and tuberous sclerosis complex (TSC) type 1 and 2 (TSC1, TSC2). Li Fraumeni Syndrome, Constitutional Mismatch Repair Deficiency, Gorlin syndrome (Nevoid Basal Cell Carcinoma), Rhabdoid Tumor Predisposition syndrome, and Von Hippel-Lindau disease. Ataxia Telangiectasia will also be discussed given the profound neurological manifestations of this syndrome. In addition, there are other cancer predisposition syndromes like Cowden/PTEN Hamartoma Tumor Syndrome, DICER1 syndrome, among many others which also increase the risk of NS neoplasia and are briefly described. Herein, we discuss the NS tumor spectrum seen in the abovementioned cancer predisposition syndromes as with their respective germline genetic abnormalities and recommended surveillance guidelines when applicable. We conclude with a discussion of the importance and rationale for genetic counseling in these patients and their families.
Collapse
Affiliation(s)
- Prabhumallikarjun Patil
- Children's Healthcare of Atlanta, Aflac Cancer Center, Atlanta, GA, USA.
- Emory University School of Medicine, Atlanta, GA, USA.
| | - Bojana Borislavova Pencheva
- Children's Healthcare of Atlanta, Aflac Cancer Center, Atlanta, GA, USA
- Emory University School of Medicine, Atlanta, GA, USA
| | - Vinayak Mahesh Patil
- Intensive Care Unit Medical Officer, District Hospital Vijayapura, Karnataka, India
| | - Jason Fangusaro
- Children's Healthcare of Atlanta, Aflac Cancer Center, Atlanta, GA, USA
- Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
23
|
Rashid S, Gupta S, McCormick SR, Tsao H. New Insights into Melanoma Tumor Syndromes. JID INNOVATIONS 2022; 2:100152. [DOI: 10.1016/j.xjidi.2022.100152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 10/14/2022] Open
|
24
|
Bruno W, Dalmasso B, Barile M, Andreotti V, Elefanti L, Colombino M, Vanni I, Allavena E, Barbero F, Passoni E, Merelli B, Pellegrini S, Morgese F, Danesi R, Calò V, Bazan V, D'Elia AV, Molica C, Gensini F, Sala E, Uliana V, Soma PF, Genuardi M, Ballestrero A, Spagnolo F, Tanda E, Queirolo P, Mandalà M, Stanganelli I, Palmieri G, Menin C, Pastorino L, Ghiorzo P. Predictors of germline status for hereditary melanoma: 5 years of multi-gene panel testing within the Italian Melanoma Intergroup. ESMO Open 2022; 7:100525. [PMID: 35777164 PMCID: PMC9434136 DOI: 10.1016/j.esmoop.2022.100525] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/26/2022] [Accepted: 05/29/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND The incidence of cutaneous melanoma is increasing in Italy, in parallel with the implementation of gene panels. Therefore, a revision of national genetic assessment criteria for hereditary melanoma may be needed. The aim of this study was to identify predictors of susceptibility variants in the largest prospective cohort of Italian high-risk melanoma cases studied to date. MATERIALS AND METHODS From 25 Italian centers, we recruited 1044 family members and germline sequenced 940 cutaneous melanoma index cases through a shared gene panel, which included the following genes: CDKN2A, CDK4, BAP1, POT1, ACD, TERF2IP, MITF and ATM. We assessed detection rate according to familial status, region of origin, number of melanomas and presence and type of non-melanoma tumors. RESULTS The overall detection rate was 9.47% (5.53% analyzing CDKN2A alone), ranging from 5.14% in sporadic multiple melanoma cases (spoMPM) with two cutaneous melanomas to 13.9% in familial cases with at least three affected members. Three or more cutaneous melanomas in spoMPM cases, pancreatic cancer and region of origin predicted germline status [odds ratio (OR) = 3.23, 3.15, 2.43, P < 0.05]. Conversely, age > 60 years was a negative independent predictor (OR = 0.13, P = 0.008), and was the age category with the lowest detection rate, especially for CDKN2A. Detection rate was 19% when cutaneous melanoma and pancreatic cancer clustered together. CONCLUSIONS Gene panel doubled the detection rate given by CDKN2A alone. National genetic testing criteria may need a revision, especially regarding age cut-off (60) in the absence of strong family history, pancreatic cancer and/or a high number of cutaneous melanomas.
Collapse
Affiliation(s)
- W Bruno
- IRCCS Ospedale Policlinico San Martino, Genetics of Rare Cancers, Genoa, Italy; University of Genoa, Department of Internal Medicine and Medical Specialties (DiMI), Genoa, Italy.
| | - B Dalmasso
- IRCCS Ospedale Policlinico San Martino, Genetics of Rare Cancers, Genoa, Italy
| | - M Barile
- IRCCS Ospedale Policlinico San Martino, Genetics of Rare Cancers, Genoa, Italy
| | - V Andreotti
- IRCCS Ospedale Policlinico San Martino, Genetics of Rare Cancers, Genoa, Italy
| | - L Elefanti
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - M Colombino
- Unit of Cancer Genetics, Institute of Genetics and Biomedical Research of the National Research Council (IRGB-CNR), Sassari, Italy
| | - I Vanni
- IRCCS Ospedale Policlinico San Martino, Genetics of Rare Cancers, Genoa, Italy; University of Genoa, Department of Internal Medicine and Medical Specialties (DiMI), Genoa, Italy
| | - E Allavena
- University of Genoa, Department of Internal Medicine and Medical Specialties (DiMI), Genoa, Italy
| | - F Barbero
- IRCCS Ospedale Policlinico San Martino, Genetics of Rare Cancers, Genoa, Italy
| | - E Passoni
- Dermatology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - B Merelli
- Oncology Unit, ASST Papa Giovanni XXIIII, Bergamo, Italy
| | - S Pellegrini
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padua, Padua, Italy
| | - F Morgese
- Oncology Unit, AOU Ospedali Riuniti di Ancona, Ancona, Italy
| | - R Danesi
- Romagna Cancer Registry, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) 'Dino Amadori', Meldola, Italy
| | - V Calò
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - V Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - A V D'Elia
- Institute of Medical Genetics, ASUFC University Hospital of Udine, Udine, Italy
| | - C Molica
- Medical Oncology Unit, S. Maria della Misericordia Hospital, Perugia, Italy
| | - F Gensini
- Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - E Sala
- Cytogenetics and Medical Genetics Unit, H San Gerardo ASST Monza, Monza, Italy
| | - V Uliana
- Medical Genetics Unit, AOU di Parma, Parma, Italy
| | - P F Soma
- Casa di Cura Gibiino, Catania, Italy
| | - M Genuardi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Medical Genetics Unit, Rome, Italy; Università Cattolica del Sacro Cuore, Department of Life Sciences and Public Health, Rome, Italy
| | - A Ballestrero
- IRCCS Ospedale Policlinico San Martino, Genetics of Rare Cancers, Genoa, Italy; University of Genoa, Department of Internal Medicine and Medical Specialties (DiMI), Genoa, Italy
| | - F Spagnolo
- IRCCS Ospedale Policlinico San Martino, Medical Oncology 2, Genoa, Italy
| | - E Tanda
- IRCCS Ospedale Policlinico San Martino, Medical Oncology 2, Genoa, Italy
| | - P Queirolo
- Division of Medical Oncology for Melanoma, Sarcoma, and Rare Tumors, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - M Mandalà
- Medical Oncology Unit, S. Maria della Misericordia Hospital, Perugia, Italy; Department of Surgery and Medicine, University of Perugia, Perugia, Italy
| | - I Stanganelli
- Skin Cancer Unit, IRCCS IRST Istituto Scientifico Romagnolo per lo Studio dei Tumori 'Dino Amadori' (IRST) IRCCS, Meldola, Italy; Dermatologic Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - G Palmieri
- Unit of Cancer Genetics, Institute of Genetics and Biomedical Research of the National Research Council (IRGB-CNR), Sassari, Italy
| | - C Menin
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - L Pastorino
- IRCCS Ospedale Policlinico San Martino, Genetics of Rare Cancers, Genoa, Italy; University of Genoa, Department of Internal Medicine and Medical Specialties (DiMI), Genoa, Italy
| | - P Ghiorzo
- IRCCS Ospedale Policlinico San Martino, Genetics of Rare Cancers, Genoa, Italy; University of Genoa, Department of Internal Medicine and Medical Specialties (DiMI), Genoa, Italy
| |
Collapse
|
25
|
Giordo R, Gulsha R, Kalla S, Calin GA, Lipovich L. LncRNA-Associated Genetic Etiologies Are Shared between Type 2 Diabetes and Cancers in the UAE Population. Cancers (Basel) 2022; 14:3313. [PMID: 35884374 PMCID: PMC9313416 DOI: 10.3390/cancers14143313] [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: 03/16/2022] [Revised: 04/22/2022] [Accepted: 05/06/2022] [Indexed: 12/13/2022] Open
Abstract
Numerous epidemiological studies place patients with T2D at a higher risk for cancer. Many risk factors, such as obesity, ageing, poor diet and low physical activity, are shared between T2D and cancer; however, the biological mechanisms linking the two diseases remain largely unknown. The advent of genome wide association studies (GWAS) revealed large numbers of genetic variants associated with both T2D and cancer. Most significant disease-associated variants reside in non-coding regions of the genome. Several studies show that single nucleotide polymorphisms (SNPs) at or near long non-coding RNA (lncRNA) genes may impact the susceptibility to T2D and cancer. Therefore, the identification of genetic variants predisposing individuals to both T2D and cancer may help explain the increased risk of cancer in T2D patients. We aim to investigate whether lncRNA genetic variants with significant diabetes and cancer associations overlap in the UAE population. We first performed an annotation-based analysis of UAE T2D GWAS, confirming the high prevalence of variants at or near non-coding RNA genes. We then explored whether these T2D SNPs in lncRNAs were relevant to cancer. We highlighted six non-coding genetic variants, jointly reaching statistical significance in T2D and cancer, implicating a shared genetic architecture between the two diseases in the UAE population.
Collapse
Affiliation(s)
- Roberta Giordo
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates; (R.G.); (R.G.); (S.K.)
| | - Rida Gulsha
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates; (R.G.); (R.G.); (S.K.)
| | - Sarah Kalla
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates; (R.G.); (R.G.); (S.K.)
| | - George A. Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Leonard Lipovich
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates; (R.G.); (R.G.); (S.K.)
| |
Collapse
|
26
|
A Genome-First Approach to Estimate Prevalence of Germline Pathogenic Variants and Risk of Pancreatic Cancer in Select Cancer Susceptibility Genes. Cancers (Basel) 2022; 14:cancers14133257. [PMID: 35805029 PMCID: PMC9265005 DOI: 10.3390/cancers14133257] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 12/20/2022] Open
Abstract
Patients with germline pathogenic variants (GPV) in cancer predisposition genes are at increased risk of pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer. The genes most frequently found to harbor GPV in unselected PDAC cases are ATM, BRCA1, BRCA2, CDKN2A, CHEK2, and PALB2. However, GPV prevalence and gene-specific associations have not been extensively studied in the general population. To further explore these associations, we analyzed genomic and phenotypic data obtained from the UK Biobank (UKB) and Geisinger MyCode Community Health Initiative (GHS) cohorts comprising 200,600 and 175,449 participants, respectively. We estimated the frequency and calculated relative risks (RRs) of heterozygotes in both cohorts and a subset of individuals with PDAC. The combined frequency of heterozygous carriers of GPV in the general population ranged from 1.22% for CHEK2 to 0.05% for CDKN2A. The frequency of GPV in PDAC cases varied from 2.38% (ATM) to 0.19% (BRCA1 and CDKN2A). The RRs of PDAC were elevated for all genes except for BRCA1 and varied widely by gene from high (ATM) to low (CHEK2, BRCA2). This work expands our understanding of the frequencies of GPV heterozygous carriers and associations between PDAC and GPV in several important PDAC susceptibility genes.
Collapse
|
27
|
Abstract
Background It is estimated that about 10% of pancreatic cancer cases have a genetic background. People with a familial predisposition to pancreatic cancer can be divided into 2 groups. The first is termed hereditary pancreatic cancer, which occurs in individuals with a known hereditary cancer syndrome caused by germline single gene mutations (e.g., BRCA1/2, CDKN2A). The second is considered as familial pancreatic cancer, which is associated with several genetic factors responsible for the more common development of pancreatic cancer in certain families, but the precise single gene mutation has not been found. Aim This review summarizes the current state of knowledge regarding the risk of pancreatic cancer development in hereditary pancreatic cancer and familial pancreatic cancer patients. Furthermore, it gathers the latest recommendations from the three major organizations dealing with the prevention of pancreatic cancer in high-risk groups and explores recent guidelines of scientific societies on screening for pancreatic cancers in individuals at risk for hereditary or familial pancreatic cancer. Conclusions In order to improve patients’ outcomes, authors of current guidelines recommend early and intensive screening in patients with pancreatic cancer resulting from genetic background. The screening should be performed in excellence centers. The scope, extent and cost-effectiveness of such interventions requires further studies.
Collapse
|
28
|
Perez‐Becerril C, Wallace AJ, Schlecht H, Bowers NL, Smith PT, Gokhale C, Eaton H, Charlton C, Robinson R, Charlton RS, Evans DG, Smith MJ. Screening of potential novel candidate genes in schwannomatosis patients. Hum Mutat 2022; 43:1368-1376. [PMID: 35723634 PMCID: PMC9540472 DOI: 10.1002/humu.24424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 06/10/2022] [Accepted: 06/15/2022] [Indexed: 01/07/2023]
Abstract
Schwannomatosis comprises a group of hereditary tumor predisposition syndromes characterized by, usually benign, multiple nerve sheath tumors, which frequently cause severe pain that does not typically respond to drug treatments. The most common schwannomatosis‐associated gene is NF2, but SMARCB1 and LZTR1 are also associated. There are still many cases in which no pathogenic variants (PVs) have been identified, suggesting the existence of as yet unidentified genetic risk factors. In this study, we performed extended genetic screening of 75 unrelated schwannomatosis patients without identified germline PVs in NF2, LZTR1, or SMARCB1. Screening of the coding region of DGCR8, COQ6, CDKN2A, and CDKN2B was carried out, based on previous reports that point to these genes as potential candidate genes for schwannomatosis. Deletions or duplications in CDKN2A, CDKN2B, and adjacent chromosome 9 region were assessed by multiplex ligation‐dependent probe amplification analysis. Sequencing analysis of a patient with multiple schwannomas and melanomas identified a novel duplication in the coding region of CDKN2A, disrupting both p14ARF and p16INK4a. Our results suggest that none of these genes are major contributors to schwannomatosis risk but the possibility remains that they may have a role in more complex mechanisms for tumor predisposition.
Collapse
Affiliation(s)
- Cristina Perez‐Becerril
- School of Biological Sciences, Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
- Manchester Centre for Genomic Medicine, St Mary's HospitalManchester University NHS Foundation TrustManchesterUK
| | - Andrew J. Wallace
- Manchester Centre for Genomic Medicine, St Mary's HospitalManchester University NHS Foundation TrustManchesterUK
| | - Helene Schlecht
- Manchester Centre for Genomic Medicine, St Mary's HospitalManchester University NHS Foundation TrustManchesterUK
| | - Naomi L. Bowers
- Manchester Centre for Genomic Medicine, St Mary's HospitalManchester University NHS Foundation TrustManchesterUK
| | - Philip T. Smith
- Manchester Centre for Genomic Medicine, St Mary's HospitalManchester University NHS Foundation TrustManchesterUK
| | - Carolyn Gokhale
- Manchester Centre for Genomic Medicine, St Mary's HospitalManchester University NHS Foundation TrustManchesterUK
| | - Helen Eaton
- Manchester Centre for Genomic Medicine, St Mary's HospitalManchester University NHS Foundation TrustManchesterUK
| | - Chris Charlton
- Manchester Centre for Genomic Medicine, St Mary's HospitalManchester University NHS Foundation TrustManchesterUK
| | - Rachel Robinson
- North East and Yorkshire Genomic Laboratory HubSt James's University HospitalLeedsUK
| | - Ruth S. Charlton
- North East and Yorkshire Genomic Laboratory HubSt James's University HospitalLeedsUK
| | - D. Gareth Evans
- School of Biological Sciences, Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
- Manchester Centre for Genomic Medicine, St Mary's HospitalManchester University NHS Foundation TrustManchesterUK
| | - Miriam J. Smith
- School of Biological Sciences, Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
- Manchester Centre for Genomic Medicine, St Mary's HospitalManchester University NHS Foundation TrustManchesterUK
| |
Collapse
|
29
|
Klatte DCF, Wallace MB, Löhr M, Bruno MJ, van Leerdam ME. Hereditary pancreatic cancer. Best Pract Res Clin Gastroenterol 2022; 58-59:101783. [PMID: 35988957 DOI: 10.1016/j.bpg.2021.101783] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/09/2021] [Accepted: 12/15/2021] [Indexed: 01/31/2023]
Abstract
Pancreatic cancer is one of the deadliest malignancies. Therefore, there is an urgent need to detect pancreatic cancer in an earlier stage to improve outcomes. A variety of hereditary cancer syndromes have been associated with an increased risk of developing pancreatic cancer, and these individuals may benefit from surveillance programs. Surveillance programs have shown potential to improve outcomes, but have important risks such as overtreatment. In this review we will discuss the definitions and epidemiology of hereditary pancreatic cancer, recommendations for genetic testing and participation in surveillance. Important aspects are differences in surveillance strategies, target lesions, and potential benefits and harms of surveillance. Lastly we will highlight future directions for research and improvement of care for individuals at high-risk of pancreatic cancer.
Collapse
Affiliation(s)
- Derk C F Klatte
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands; Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, United States.
| | - Michael B Wallace
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, United States; Division of Gastroenterology and Hepatology, Sheikh Shakhbout Medical City, Abu Dhabi, United Arab Emirates.
| | - Matthias Löhr
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | - Marco J Bruno
- Department of Gastroenterology and Hepatology, Erasmus Medical Center, Rotterdam, the Netherlands.
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands; Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
30
|
Arnold M, Singh D, Laversanne M, Vignat J, Vaccarella S, Meheus F, Cust AE, de Vries E, Whiteman DC, Bray F. Global Burden of Cutaneous Melanoma in 2020 and Projections to 2040. JAMA Dermatol 2022; 158:495-503. [PMID: 35353115 PMCID: PMC8968696 DOI: 10.1001/jamadermatol.2022.0160] [Citation(s) in RCA: 554] [Impact Index Per Article: 184.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/16/2022] [Indexed: 12/15/2022]
Abstract
Importance Despite many cases being preventable, cutaneous melanoma remains the most serious skin cancer worldwide. Understanding the scale and profile of the disease is vital to concentrate and reinforce global prevention efforts. Objective To examine global patterns of cutaneous melanoma in 2020 and to provide projected estimates of cases and deaths by 2040. Design, Setting, and Participants This population-based study used the GLOBOCAN 2020 database for global epidemiological assessment of new cases and deaths due to invasive melanoma. Main Outcomes and Measures Age-standardized incidence and mortality rates were calculated per 100 000 person-years by country, world region, and 4-tier level of human development. Estimated numbers of cases and deaths were calculated for the year 2040. Results A worldwide total of 325 000 new melanoma cases (174 000 males, 151 000 females) and 57 000 deaths (32 000 males, 25 000 females) was estimated for 2020. Large geographic variations existed across countries and world regions, with the highest incidence rates among males (42 per 100 000 person-years) and females (31 per 100 000 person-years) observed in Australia/New Zealand, followed by Western Europe (19 per 100 000 person-years for males and females), North America (18 per 100 000 person-years for males, 14 per 100 000 person-years for females), and Northern Europe (17 per 100 000 person-years for males, 18 per 100 000 person-years for females). Melanoma continued to be rare in most African and Asian countries, with incidence rates commonly less than 1 per 100 000 person-years. Mortality rates peaked at 5 per 100 000 person-years in New Zealand, and geographic variations were less pronounced than for incidence. Melanoma was more frequent among males than females in most world regions. If 2020 rates continue, the burden from melanoma is estimated to increase to 510 000 new cases (a roughly 50% increase) and to 96 000 deaths (a 68% increase) by 2040. Conclusions and Relevance This epidemiological assessment suggests that melanoma remains an important challenge to cancer control and public health globally, especially in fair-skinned populations of European descent.
Collapse
Affiliation(s)
- Melina Arnold
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Deependra Singh
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Mathieu Laversanne
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Jerome Vignat
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Salvatore Vaccarella
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Filip Meheus
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Anne E. Cust
- The Daffodil Centre, The University of Sydney with Cancer Council New South Wales, Sydney, Australia
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Esther de Vries
- Department of Clinical Epidemiology and Biostatistics, Pontificia Universidad Javeriana, Bogota, Colombia
| | - David C. Whiteman
- Cancer Control Group, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Freddie Bray
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| |
Collapse
|
31
|
Soewito S, Wyatt R, Berenson E, Poullard N, Gessay S, Mette L, Marin E, Shelby K, Alvarez E, Choi BY, Aviles C, Pulido-Saldivar AM, Otto PM, Jatoi I, Ramamurthy C, Ignatius M, Kaklamani VG, Tomlinson GE. Disparities in Cancer Genetic Testing and Variants of Uncertain Significance in the Hispanic Population of South Texas. JCO Oncol Pract 2022; 18:e805-e813. [PMID: 35544645 PMCID: PMC10166383 DOI: 10.1200/op.22.00090] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Racial and ethnic disparities have included a lack of access to both genetic testing and research, resulting in poor understanding of the genomic architecture in under-represented populations. The South Texas population is primarily of Hispanic background and has been largely devoid of genetic services. We extended access to this underserved population and uncovered genetic variants previously not observed, emphasizing the need to continually improve both genomic databases and clarification of variant significance to provide meaningful patient counseling. METHODS This study consisted of a retrospective cohort review of patients seen through a cancer genetics education and service program across 24 counties in South Texas. In total, 1,595 individuals were identified as appropriate for cancer genetic counseling and 1,377 completed genetic testing. RESULTS Eighty percent of those receiving genetic counseling self-identified as Hispanic, 16% as non-Hispanic White (NHW), 3% as African American, and 1% as other race/ethnicity. Of reported variants, 18.8% were pathogenic and 13.7% were reported as a variant of uncertain significance (VUS). VUS was reported in 17.2% of the Hispanic individuals compared with 9% NHW (P = .005). CONCLUSION Individuals of Hispanic ethnicity were significantly more likely to harbor a VUS compared with NHW. The extended reach into our regional communities revealed a gap in the ability to accurately interpret genomic variation with implications for advising patients on screening, prevention, and management strategies. A higher percentage of VUS also emphasizes the challenge of continued follow-up amid existing barriers that led to disparities in access. As understanding of the variants develops, hopefully gaps in knowledge of the genomic landscape will be lessened with increased clarity to provide accurate cancer risk assessment and recommendations for implementing prevention initiatives.
Collapse
Affiliation(s)
| | - Rachel Wyatt
- Department of Pediatrics, UT Health San Antonio, San Antonio, TX
| | - Emily Berenson
- Department of Pediatrics, UT Health San Antonio, San Antonio, TX
| | | | - Shawn Gessay
- Department of Pediatrics, UT Health San Antonio, San Antonio, TX
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX
- Present address: PreventionGenetics, Marshfield, WI
| | - Lindsey Mette
- Department of Pediatrics, UT Health San Antonio, San Antonio, TX
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX
- Present address: Invitae, San Francisco, CA
| | - Elena Marin
- Greehey Children's Cancer Research Institute, UT Health San Antonio, TX
| | - Kristin Shelby
- Greehey Children's Cancer Research Institute, UT Health San Antonio, TX
| | - Elise Alvarez
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX
| | - Byeong Yeob Choi
- Department of Population Health Sciences, UT Health San Antonio TX, San Antonio, TX
| | - Clarissa Aviles
- Greehey Children's Cancer Research Institute, UT Health San Antonio, TX
| | - Anna Maria Pulido-Saldivar
- Greehey Children's Cancer Research Institute, UT Health San Antonio, TX
- UT Laredo Campus, UT Health San Antonio, Laredo, TX
| | - Pamela M. Otto
- Department of Radiology, UT Health San Antonio, San Antonio, TX
| | - Ismail Jatoi
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX
- Department of Surgical Oncology, UT Health San Antonio, San Antonio, TX
| | | | - Myron Ignatius
- Greehey Children's Cancer Research Institute, UT Health San Antonio, TX
| | | | - Gail E. Tomlinson
- Department of Pediatrics, UT Health San Antonio, San Antonio, TX
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX
- Greehey Children's Cancer Research Institute, UT Health San Antonio, TX
| |
Collapse
|
32
|
Pauley K, Khan A, Kohlmann W, Jeter J. Considerations for Germline Testing in Melanoma: Updates in Behavioral Change and Pancreatic Surveillance for Carriers of CDKN2A Pathogenic Variants. Front Oncol 2022; 12:837057. [PMID: 35372037 PMCID: PMC8967159 DOI: 10.3389/fonc.2022.837057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/23/2022] [Indexed: 01/13/2023] Open
Abstract
The largest proportion of hereditary melanoma cases are due to pathogenic variants (PVs) in the CDKN2A/p16 gene, which account for 20%-40% of familial melanomas and confer up to a 30%-70% lifetime risk for melanoma in individuals with these variants. In addition, PVs in the CDKN2A gene also increase risk for pancreatic cancer (~5-24% lifetime risk). Individuals with PVs in the CDKN2A gene also tend to have an earlier onset of cancer. Despite these known risks, uptake of germline testing has been limited in the past, largely due to perceptions of limited benefit for patients. Prevention recommendations have been developed for individuals with CDKN2A PVs as well the providers who care for them. On the patient level, behavioral modifications regarding melanoma prevention such as wearing sunscreen, limiting prolonged sun exposure and practicing general sun safety can help reduce risks. Germline testing can provide motivation for some individuals to adhere to these lifestyle changes. On the provider level, pancreatic cancer surveillance for individuals with CDKN2A PVs has been increasingly endorsed by expert consensus, although the efficacy of these surveillance methods remains under study. This review summarizes the updated surveillance guidelines for individuals with CDKN2A PVs and explores the impact of genetic counseling and testing in influencing behavioral changes in these individuals.
Collapse
Affiliation(s)
- Kristen Pauley
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, Salt Lake City, UT, United States
| | - Ambreen Khan
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, Salt Lake City, UT, United States
| | - Wendy Kohlmann
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, Salt Lake City, UT, United States
| | - Joanne Jeter
- Department of Internal Medicine, Huntsman Cancer Institute, Salt Lake City, UT, United States
| |
Collapse
|
33
|
Jeong AR, Forbes K, Orosco RK, Cohen EEW. Hereditary oral squamous cell carcinoma associated with CDKN2A germline mutation: a case report. J Otolaryngol Head Neck Surg 2022; 51:5. [PMID: 35123577 PMCID: PMC8818223 DOI: 10.1186/s40463-022-00556-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 01/02/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Germline CDKN2A mutations are a well-known cause of familial atypical multiple mole melanoma (OMIM #155601) and melanoma-pancreatic cancer syndrome (OMIM #606719). Increased risk of head and neck squamous cell carcinoma (HNSCC), particularly oral squamous cell carcinoma (OSCC) in those with germline CDKN2A mutations, has been described. However, screening for HNSCC is not a routine practice in patients with CDKN2A germline mutations and these mutations are not a conventional test for HNSCC patients without obvious risk factors. CASE PRESENTATION We describe a female with no smoking history who developed oral squamous cell carcinoma at age 39 and had a complex clinical course of recurrent multifocal squamous cell carcinoma (SCC) and carcinoma in situ of the oral cavity and oropharynx. Detailed family history demonstrated that her mother was diagnosed with OSCC and melanoma in her 40 s, and her maternal grandfather was diagnosed with metastatic melanoma in his 40 s. Genetic testing of the patient and her mother revealed CDKN2A c.301G>T mutation. She was referred to genetic counseling as well as to dermatology, gastroenterology, and neurology for cancer surveillance. She was treated with resections and has no evidence of disease 3 years after diagnosis. CONCLUSIONS We report a family with a CDKN2A c.301 G>T mutation who also have significant history of OSCC, adding to the growing body of literature suggesting increased risk of HNSCC, particularly OSCC, in CDKN2A germline mutation carriers. It is important to consider CDKN2A mutation testing in familial HNSCC and young patients without obvious risk factors. Moreover, surveillance for HNSCC should be routine practice in those with a CDKN2A germline mutation.
Collapse
Affiliation(s)
- Ah-Reum Jeong
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego, 3855 Health Sciences Drive, La Jolla, CA, 92093-0960, USA
| | - Kimberly Forbes
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego, 3855 Health Sciences Drive, La Jolla, CA, 92093-0960, USA
| | - Ryan K Orosco
- Division of Otolaryngology, Department of Surgery, University of California San Diego, La Jolla, CA, 92093, USA.,Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ezra E W Cohen
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego, 3855 Health Sciences Drive, La Jolla, CA, 92093-0960, USA. .,Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
34
|
Chen Z, Guo Y, Zhao D, Zou Q, Yu F, Zhang L, Xu L. Comprehensive Analysis Revealed that CDKN2A is a Biomarker for Immune Infiltrates in Multiple Cancers. Front Cell Dev Biol 2022; 9:808208. [PMID: 35004697 PMCID: PMC8733648 DOI: 10.3389/fcell.2021.808208] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/06/2021] [Indexed: 01/22/2023] Open
Abstract
The CDKN2A (cyclin dependent kinase inhibitor 2A/multiple tumor suppressor 1) gene, also known as the P16 gene, encodes multiple tumor suppressor 1 (MTS1), which belongs to the INK4 family. In tumor tissue, CDKN2A has a high expression level compared with normal tissue and reflects prognosis in tumor patients. Our research targeted the analysis of CDKN2A expression in 33 tumors and clinical parameters, patient prognosis and tumor immunity roles. The CDKN2A expression level was significantly correlated with the tumor mutation burden (TMB) in 10 tumors, and the expression of CDKN2A was also correlated with MSI (microsatellite instability) in 10 tumors. CDKN2A expression was associated with infiltrating lymphocyte (TIL) levels in 22 pancancers, thus suggesting that CDKN2A expression is associated with tumor immunity. Enrichment analysis indicated that CDKN2A expression was involved in natural killer cell-mediated cytotoxicity pathways, antigen processing and presentation, olfactory transduction pathways, and regulation of the autophagy pathway in multiple cancers. CDKN2A was significantly associated with several immune cell infiltrates in pantumors. CDKN2A may serve as a promising prognostic biomarker and is associated with immune infiltrates across cancers.
Collapse
Affiliation(s)
- Zheng Chen
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China.,School of Applied Chemistry and Biological Technology, Shenzhen Polytechnic, Shenzhen, China
| | - Yingjie Guo
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China.,School of Electronic and Communication Engineering, Shenzhen Polytechnic, Shenzhen, China
| | - Da Zhao
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China.,School of Applied Chemistry and Biological Technology, Shenzhen Polytechnic, Shenzhen, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| | - Fusheng Yu
- Beidahuang Industry Group General Hospital, Harbin, China
| | - Lijun Zhang
- School of Applied Chemistry and Biological Technology, Shenzhen Polytechnic, Shenzhen, China
| | - Lei Xu
- School of Electronic and Communication Engineering, Shenzhen Polytechnic, Shenzhen, China
| |
Collapse
|
35
|
Sokolenko EA, Berchner-Pfannschmidt U, Ting SC, Schmid KW, Bechrakis NE, Seitz B, Tsimpaki T, Kraemer MM, Fiorentzis M. Optimisation of the Chicken Chorioallantoic Membrane Assay in Uveal Melanoma Research. Pharmaceutics 2021; 14:13. [PMID: 35056909 PMCID: PMC8778438 DOI: 10.3390/pharmaceutics14010013] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/04/2021] [Accepted: 12/19/2021] [Indexed: 01/07/2023] Open
Abstract
The treatment of uveal melanoma and its metastases has not evolved sufficiently over the last decades in comparison to other tumour entities, posing a great challenge in the field of ocular oncology. Despite improvements in the conventional treatment regime and new discoveries about the genetic and molecular background of the primary tumour, effective treatment strategies to either prevent tumours or treat patients with advanced or metastatic disease are still lacking. New therapeutic options are necessary in order to achieve satisfactory local tumour control, reduce the risk of metastasis development, and preserve the eyeball and possibly the visual function of the eye. The development of in vivo model systems remains crucial for the identification and investigation of potential novel treatment modalities. The aim of this study was the optimisation of the chorioallantoic membrane (CAM) model for uveal melanoma research. We analysed the established CAM assay and its modification after the implantation of three-dimensional spheroids. The chorioallantoic membrane of a chick embryo was used to implant uveal melanoma-cell-line-derived spheroids in order to study their growth rate, angiogenic potential, and metastatic capability. Using the UM 92.1, UPMD2, UPMM3, and Mel270 cell lines, we were able to improve the viability of the embryos from 20% to >80% and to achieve up to a fourfold volume increase of the transplanted spheroid masses. The results point to the value of an optimised chicken embryo assay as an in vivo model for testing novel therapies for uveal melanoma by simplifying the research conditions and by contributing to a considerable reduction in animal experiments.
Collapse
Affiliation(s)
- Ekaterina A. Sokolenko
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany; (U.B.-P.); (N.E.B.); (T.T.); (M.M.K.); (M.F.)
| | - Utta Berchner-Pfannschmidt
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany; (U.B.-P.); (N.E.B.); (T.T.); (M.M.K.); (M.F.)
| | - Saskia C. Ting
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany; (S.C.T.); (K.W.S.)
| | - Kurt W. Schmid
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany; (S.C.T.); (K.W.S.)
| | - Nikolaos E. Bechrakis
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany; (U.B.-P.); (N.E.B.); (T.T.); (M.M.K.); (M.F.)
| | - Berthold Seitz
- Department of Ophthalmology, Saarland University Medical Center, Kirrberger Str. 100, 66421 Homburg, Germany;
| | - Theodora Tsimpaki
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany; (U.B.-P.); (N.E.B.); (T.T.); (M.M.K.); (M.F.)
| | - Miriam Monika Kraemer
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany; (U.B.-P.); (N.E.B.); (T.T.); (M.M.K.); (M.F.)
| | - Miltiadis Fiorentzis
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany; (U.B.-P.); (N.E.B.); (T.T.); (M.M.K.); (M.F.)
| |
Collapse
|
36
|
Opportunities and challenges in targeted therapy and immunotherapy for pancreatic cancer. Expert Rev Mol Med 2021; 23:e21. [PMID: 34906271 DOI: 10.1017/erm.2021.26] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer is one of the most malignant tumours with a poor prognosis. In recent years, the incidence of pancreatic cancer is on the rise. Traditional chemotherapy and radiotherapy for pancreatic cancer have been improved, first-line and second-line palliative treatments have been developed, and adjuvant treatments have also been used in clinical. However, the 5-year survival rate is still less than 10% and new treatment methods such as targeted therapy and immunotherapy need to be investigated. In the past decades, many clinical trials of targeted therapies and immunotherapies for pancreatic cancer were launched and some of them showed an ideal prospect in a subgroup of pancreatic cancer patients. The experience of both success and failure of these clinical trials will be helpful to improve these therapies in the future. Therefore, the current research progress and challenges of selected targeted therapies and immunotherapies for pancreatic cancer are reviewed.
Collapse
|
37
|
Popa LG, Giurcaneanu C, Nitipir C, Popa AM, Stoica C, Beiu C, Tebeica T, Negoita S, Mihai MM. Dysplastic nevus syndrome and pancreatic cancer: A case report. Exp Ther Med 2021; 23:31. [PMID: 34824639 DOI: 10.3892/etm.2021.10953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/30/2021] [Indexed: 11/06/2022] Open
Abstract
Multiple primary cancers may occur in the same patient, with a prevalence that follows an ascendant trend. Their development is dictated by a complex interplay between a variety of factors, both patient-dependent and external. The case of a 38-year-old female patient diagnosed and treated for pancreatic cancer (PC) is presented in whom the digital dermoscopic monitoring of melanocytic nevi revealed a marked change of two nevi that acquired rapidly highly atypical features. They were surgically excised and the histopathological examination revealed two completely excised dysplastic compound nevi. Clinicians should be aware of the strong association between dysplastic nevus syndrome and PC, a malignancy associated with an extremely poor prognosis. Familial atypical multiple mole melanoma syndrome (FAMMM) predisposes to the development of melanoma, pancreatic cancer and other neoplasms. The common genetic background of PC and hereditary melanoma is discussed and the importance of regular skin checkup and screening for PC in these patients is underlined.
Collapse
Affiliation(s)
- Liliana Gabriela Popa
- Department of Dermatology, 'Elias' Emergency University Hospital, 011461 Bucharest, Romania.,Department of Oncologic Dermatology, 'Elias' Emergency University Hospital, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Calin Giurcaneanu
- Department of Dermatology, 'Elias' Emergency University Hospital, 011461 Bucharest, Romania.,Department of Oncologic Dermatology, 'Elias' Emergency University Hospital, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cornelia Nitipir
- Department of Oncology, 'Elias' Emergency University Hospital, 011461 Bucharest, Romania.,Department of Oncology, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Ana Maria Popa
- Department of Oncology, 'Elias' Emergency University Hospital, 011461 Bucharest, Romania
| | - Cristiana Stoica
- Department of Dermatology, 'Elias' Emergency University Hospital, 011461 Bucharest, Romania
| | - Cristina Beiu
- Department of Oncologic Dermatology, 'Elias' Emergency University Hospital, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Tiberiu Tebeica
- Department of Dermatopathology, 'Dr. Leventer' Centre, 011216 Bucharest, Romania
| | - Silvius Negoita
- Department of Anaesthesiology and Intensive Care, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania.,Department of Anaesthesiology and Intensive Care, 'Elias' Emergency University Hospital, 011461 Bucharest, Romania
| | - Mara Madalina Mihai
- Department of Dermatology, 'Elias' Emergency University Hospital, 011461 Bucharest, Romania.,Department of Oncologic Dermatology, 'Elias' Emergency University Hospital, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
38
|
Khaddour K, Maahs L, Avila-Rodriguez AM, Maamar Y, Samaan S, Ansstas G. Melanoma Targeted Therapies beyond BRAF-Mutant Melanoma: Potential Druggable Mutations and Novel Treatment Approaches. Cancers (Basel) 2021; 13:5847. [PMID: 34831002 PMCID: PMC8616477 DOI: 10.3390/cancers13225847] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/24/2022] Open
Abstract
Melanomas exhibit the highest rate of somatic mutations among all different types of cancers (with the exception of BCC and SCC). The accumulation of a multimode of mutations in the driver oncogenes are responsible for the proliferative, invasive, and aggressive nature of melanomas. High-resolution and high-throughput technology has led to the identification of distinct mutational signatures and their downstream alterations in several key pathways that contribute to melanomagenesis. This has enabled the development of individualized treatments by targeting specific molecular alterations that are vital for cancer cell survival, which has resulted in improved outcomes in several cancers, including melanomas. To date, BRAF and MEK inhibitors remain the only approved targeted therapy with a high level of evidence in BRAFV600E/K mutant melanomas. The lack of approved precision drugs in melanomas, relative to other cancers, despite harboring one of the highest rates of somatic mutations, advocates for further research to unveil effective therapeutics. In this review, we will discuss potential druggable mutations and the ongoing research of novel individualized treatment approaches targeting non-BRAF mutations in melanomas.
Collapse
Affiliation(s)
- Karam Khaddour
- Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (L.M.); (A.M.A.-R.)
| | - Lucas Maahs
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (L.M.); (A.M.A.-R.)
| | - Ana Maria Avila-Rodriguez
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (L.M.); (A.M.A.-R.)
| | - Yazan Maamar
- Division of Hematology and Oncology, Department of Medicine, University of Tishreen Lattakia, Lattakia 2217, Syria;
| | - Sami Samaan
- Department of Medicine, American University of Beirut, Beirut 1107, Lebanon;
| | - George Ansstas
- Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| |
Collapse
|
39
|
Potjer TP, van der Grinten TWJ, Lakeman IMM, Bollen SH, Rodríguez-Girondo M, Iles MM, Barrett JH, Kiemeney LA, Gruis NA, van Asperen CJ, van der Stoep N. Association between a 46-SNP Polygenic Risk Score and melanoma risk in Dutch patients with familial melanoma. J Med Genet 2021; 58:760-766. [PMID: 32994281 PMCID: PMC8551976 DOI: 10.1136/jmedgenet-2020-107251] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/13/2020] [Accepted: 08/16/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND Familial clustering of melanoma suggests a shared genetic predisposition among family members, but only 10%-40% of familial cases carry a pathogenic variant in a known high-risk melanoma susceptibility gene. We investigated whether a melanoma-specific Polygenic Risk Score (PRS) is associated with melanoma risk in patients with genetically unexplained familial melanoma. METHODS Dutch familial melanoma cases (n=418) were genotyped for 46 SNPs previously identified as independently associated with melanoma risk. The 46-SNP PRS was calculated and standardised to 3423 healthy controls (sPRS) and the association between PRS and melanoma risk was modelled using logistic regression. Within the case series, possible differences were further explored by investigating the PRS in relation to (1) the number of primary melanomas in a patient and (2) the extent of familial clustering of melanoma. RESULTS The PRS was significantly associated with melanoma risk, with a per-SD OR of 2.12 (95% CI 1.90 to 2.35, p<0.001), corresponding to a 5.70-fold increased risk (95% CI 3.93 to 8.28) when comparing the top 90th to the middle 40-60th PRS percentiles. The mean PRS was significantly higher in cases with multiple primary melanomas than in cases with a single melanoma (sPRS 1.17 vs 0.71, p=0.001). Conversely, cases from high-density melanoma families had a lower (but non-significant) mean PRS than cases from low-density families (sPRS 0.60 vs 0.94, p=0.204). CONCLUSION Our work underlines the significance of a PRS in determining melanoma susceptibility and encourages further exploration of the diagnostic value of a PRS in genetically unexplained melanoma families.
Collapse
Affiliation(s)
- Thomas P Potjer
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Inge M M Lakeman
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Sander H Bollen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Mar Rodríguez-Girondo
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Mark M Iles
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, Leeds, UK
| | - Jennifer H Barrett
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, Leeds, UK
| | - Lambertus A Kiemeney
- Department of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nelleke A Gruis
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Christi J van Asperen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Nienke van der Stoep
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
40
|
CDKN2A-Mutated Pancreatic Ductal Organoids from Induced Pluripotent Stem Cells to Model a Cancer Predisposition Syndrome. Cancers (Basel) 2021; 13:cancers13205139. [PMID: 34680288 PMCID: PMC8533699 DOI: 10.3390/cancers13205139] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/05/2021] [Accepted: 10/09/2021] [Indexed: 12/20/2022] Open
Abstract
Patient-derived induced pluripotent stem cells (iPSCs) provide a unique platform to study hereditary disorders and predisposition syndromes by resembling germline mutations of affected individuals and by their potential to differentiate into nearly every cell type of the human body. We employed plucked human hair from two siblings with a family history of cancer carrying a pathogenic CDKN2A variant, P16-p.G101W/P14-p.R115L, to generate patient-specific iPSCs in a cancer-prone ancestry for downstream analytics. The differentiation capacity to pancreatic progenitors and to pancreatic duct-like organoids (PDLOs) according to a recently developed protocol remained unaffected. Upon inducible expression of KRASG12Dusing a piggyBac transposon system in CDKN2A-mutated PDLOs, we revealed structural and molecular changes in vitro, including disturbed polarity and epithelial-to-mesenchymal (EMT) transition. CDKN2A-mutated KRASG12DPDLO xenotransplants formed either a high-grade precancer lesion or a partially dedifferentiated PDAC-like tumor. Intriguingly, P14/P53/P21 and P16/RB cell-cycle checkpoint controls have been only partly overcome in these grafts, thereby still restricting the tumorous growth. Hereby, we provide a model for hereditary human pancreatic cancer that enables dissection of tumor initiation and early development starting from patient-specific CDKN2A-mutated pluripotent stem cells.
Collapse
|
41
|
Kimura H, Klein AP, Hruban RH, Roberts NJ. The Role of Inherited Pathogenic CDKN2A Variants in Susceptibility to Pancreatic Cancer. Pancreas 2021; 50:1123-1130. [PMID: 34714275 PMCID: PMC8562885 DOI: 10.1097/mpa.0000000000001888] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
ABSTRACT CDKN2A is cell cycle negative regulator, and the role of CDKN2A in the development of pancreatic ductal adenocarcinoma, which continues to be a lethal cancer, is well-established. Somatic loss of CDKN2A is considered one of the major drivers of pancreatic tumorigenesis. CDKN2A gene is one of the pancreatic cancer susceptibility gene; in addition to melanoma, pathogenic germline CDKN2A variants have been identified in up to 3.3% patients with pancreatic ductal adenocarcinoma depending on family history of disease. Carriers of a known pathogenic germline CDKN2A variant have up to a 12.3-fold increased risk of developing pancreatic cancer. Recently, several studies have demonstrated the benefit of clinical surveillance in patients with pathogenic germline CDKN2A variants. Therefore, identification of patients with a pathogenic germline CDKN2A variant is important for screening of at-risk relatives for pancreatic cancer. It has the potential to lead to the detection of early, potentially curable pancreatic cancer and precursor neoplasms, and reduce mortality. Furthermore, patients with a germline pathogenic CDKN2A variant and somatic loss of CDKN2A may benefit in the future from treatment with targeted therapies, such as a CDK4/6 inhibitor.
Collapse
Affiliation(s)
- Hirokazu Kimura
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alison P. Klein
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Epidemiology, the Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Ralph H. Hruban
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nicholas J. Roberts
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
42
|
Familial Melanoma and Susceptibility Genes: A Review of the Most Common Clinical and Dermoscopic Phenotypic Aspect, Associated Malignancies and Practical Tips for Management. J Clin Med 2021; 10:jcm10163760. [PMID: 34442055 PMCID: PMC8397216 DOI: 10.3390/jcm10163760] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/13/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022] Open
Abstract
A family history of melanoma greatly increases the risk of developing cutaneous melanoma, a highly aggressive skin cancer whose incidence has been steadily increasing worldwide. Familial melanomas account for about 10% of all malignant melanomas and display an inheritance pattern consistent with the presence of pathogenic germline mutations, among which those involving CDKN2A are the best characterized. In recent years, a growing number of genes, such as MC1R, MITF, CDK4, POT1, TERT, ACD, TERF2IP, and BAP1, have been implicated in familial melanoma. The fact that individuals harboring these germline mutations along with their close blood relatives have a higher risk of developing multiple primary melanomas as well as other internal organ malignancies, especially pancreatic cancer, makes cascade genetic testing and surveillance of these families of the utmost importance. Unfortunately, due to a polygenic inheritance mechanism involving multiple low-risk alleles, genetic modifiers, and environmental factors, it is still very difficult to predict the presence of these mutations. It is, however, known that germline mutation carriers can sometimes develop specific clinical traits, such as high atypical nevus counts and specific dermoscopic features, which could theoretically help clinicians predict the presence of these mutations in prone families. In this review, we provide a comprehensive overview of the high- and intermediate-penetrance genes primarily linked to familial melanoma, highlighting their most frequently associated non-cutaneous malignancies and clinical/dermoscopic phenotypes.
Collapse
|
43
|
Xiao M, Tang N, Yan Y, Li Z, Shi S, He S, Chen Z, Cao K, Chen J, Zhou J, Chen X. Knockdown of enhancer of rudimentary homolog expression attenuates proliferation, cell cycle and apoptosis of melanoma cells. Melanoma Res 2021; 31:309-318. [PMID: 34193803 DOI: 10.1097/cmr.0000000000000747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Early stage or localized melanoma can be surgically resected with satisfactory outcome, whereas advanced malignant melanoma responds to treatment poorly and has a negative prognosis even after surgery, radiotherapy and other comprehensive treatments. Gene therapy targeting various biological signaling pathways has become an increasingly popular area in melanoma research. However, for gene therapy success, it is important to reveal the molecular mechanisms of melanoma tumorigenesis and development. The present study examined the effects of downregulating enhancer of rudimentary homolog (ERH) expression on the proliferation, metastasis and cell cycle of melanoma cells. ERH expression levels in melanoma tissues and cells were determined. Then, ERH gene expression in melanoma cell lines was downregulated or overexpressed by the lentiviral RNA interference technique. Furthermore, we performed cell counting kit-8, clone formation, scratch, transwell migration, subcutaneous tumorigenesis and venous metastasis assays as well as carried out flow cytometry analysis to explore the effects of ERH expression on cell proliferation, cell cycle, apoptosis and metastasis. We found that ERH expression in melanoma tissues and cells was markedly higher than in normal melanin nevus. Suppressing ERH expression by RNA interference in melanoma A375, WM35 and SK28 cell lines inhibited their proliferation and induced cell apoptosis. The cell cycle was also found to be blocked in the G1 phase. However, the metastatic properties of melanoma cells in vitro and in vivo remained largely unaltered by ERH knockdown. Our results show that ERH expression is increased in melanoma. Meanwhile, the proliferation and cell cycle transformation abilities are impaired potentially by downregulating the ERH expression in melanoma cells. Therefore, targeting ERH might serve as a novel therapeutic approach for malignant melanoma.
Collapse
Affiliation(s)
- Muzhang Xiao
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Changsha
- Xiangya Changde Hospital, Changde
| | - Ningning Tang
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Changsha
- Xiangya Changde Hospital, Changde
| | - Yu Yan
- Department of Plastic surgery
| | | | | | - Siqi He
- Department of Plastic surgery
| | | | - Ke Cao
- Department of Oncology, The Third Xiangya Hospital, Changsha
| | | | | | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Changsha, Hunan, People's Republic of China. Changsha, Hunan, China
| |
Collapse
|
44
|
Abstract
Melanoma accounts for approximately 1% of all skin cancers but contributes to almost all skin cancer deaths. The developing picture suggests that melanoma phenotypes are driven by epigenetic mechanisms that reflect a complex interplay between genotype and environment. Furthermore, the growing consensus is that current classification standards, notwithstanding pertinent clinical history and appropriate biopsy, fall short of capturing the vast complexity of the disease. This article summarizes the current understanding of the clinical picture of melanoma, with a focus on the tremendous breakthroughs in molecular classification and therapeutics.
Collapse
Affiliation(s)
- Sarem Rashid
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02466, USA; Boston University School of Medicine, Boston, MA, USA
| | - Hensin Tsao
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02466, USA.
| |
Collapse
|
45
|
Pissa M, Helkkula T, Appelqvist F, Silander G, Borg Å, Pettersson J, Lapins J, Nielsen K, Höiom V, Helgadottir H. CDKN2A genetic testing in melanoma-prone families in Sweden in the years 2015-2020: implications for novel national recommendations. Acta Oncol 2021; 60:888-896. [PMID: 33945383 DOI: 10.1080/0284186x.2021.1914346] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Background: Inherited pathogenic variants (PVs) in the CDKN2A gene are among the strongest known risk factors for cutaneous melanoma. Carriers are at high risks to develop multiple primary melanomas and other cancers, in particular pancreatic cancer. In this study, the CDKN2A testing, carried out in Sweden in the years 2015-2020, was evaluated.Materials and methods: Included families had (1) three or more cases of melanoma and/or pancreatic cancer, (2) two melanomas in first-degree relatives, the youngest case <55 years or (3) individuals with three or more multiple primary melanomas, the first before the age of 55 years, and no other affected family members. The included families had at least one affected member that had been tested for CDKN2A PVs.Results: In total, 403 families were included, whereof 913 family members had been diagnosed with cutaneous melanoma and 129 with pancreatic cancer, 33 (8.2%) were found to have PVs in CDKN2A. Frequencies ranged from 0.9% in families with only two melanomas to 43.2% in families with three or more melanoma cases and pancreatic cancer (p < 0.001). The frequency of PVs ranged from 2.1% to 16.5% in families where the youngest case was ≥55 years or <35 years (p = 0.040). In families with or without CDKN2A PVs, 37.6% and 10.0% had melanoma cases that had died from melanoma, respectively (p < 0.001).Discussion: Significant differences were seen in the frequencies of CDKN2A PVs, dependent on numbers or age at diagnosis of melanomas and diagnoses of pancreatic cancers in the family. Further, melanoma cases belonging to families that tested positive for CDKN2A PVs had a significantly higher mortality. To summarize, the current evaluation shows that, with adequately selected criteria to guide genetic testing, CDKN2A PVs are identified at significant frequencies. Identification of carrier families is of importance to ensure that members are enrolled in a preventive surveillance program.
Collapse
Affiliation(s)
- Maria Pissa
- Department of Dermatology and Venereology, Ryhov County Hospital, Jönköping, Sweden
| | - Teo Helkkula
- Department of Clinical Sciences, Division of Dermatology, Lund University Skin Cancer research group, Lund University, Lund, Sweden
- Department of Dermatology, Skåne University Hospital, Lund, Sweden
| | - Frida Appelqvist
- Department of Dermatology, Institute of Clinical Sciences, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Gustav Silander
- Department of Radiation Sciences, Division of Oncology, Umeå University, Umeå, Sweden
| | - Åke Borg
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Jenny Pettersson
- Department of Oncology, Ryhov County Hospital, Jönköping, Sweden
| | - Jan Lapins
- Department of Dermatology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Kari Nielsen
- Department of Clinical Sciences, Division of Dermatology, Lund University Skin Cancer research group, Lund University, Lund, Sweden
- Department of Dermatology, Skåne University Hospital, Lund, Sweden
- Department of Dermatology, Helsingborg Hospital, Helsingborg, Sweden
| | - Veronica Höiom
- Department of Oncology and Pathology, Karolinska Institutet and Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Hildur Helgadottir
- Department of Oncology and Pathology, Karolinska Institutet and Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
46
|
Podlipnik S, Potrony M, Puig S. Genetic markers for characterization and prediction of prognosis of melanoma subtypes: a 2021 update. Ital J Dermatol Venerol 2021; 156:322-330. [PMID: 33982545 DOI: 10.23736/s2784-8671.21.06957-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In this article we examined the most important genetic markers involved in melanoma susceptibility, initiation and progression, and their impact on the prognosis of the disease. Current knowledge in melanoma genetics identifies distinct pathways to the development of different melanoma subtypes characterized by specific clinico-pathological features and partially known genetic markers, modulated by high, low or absence of cumulative sun damage. The most prevalent somatic mutations are related to the activation of the MAPK pathway, which are classified into four major subtypes: BRAF mutant, NRAS mutant, NF1 mutant and triple wild type. Moreover, germinal mutations are also involved in the characterization and predictions of prognosis in melanoma. Currently, CDKN2A is seen as the main high-risk gene involved in melanoma susceptibility being mutated in around 20% of melanoma-prone families. Other high-risk susceptibility genes described include CDK4, POT1, BAP1, TERT promoter, ACD, and TERF2IP. Melanoma is one of the most genetically predisposed among all cancers in humans, and ultraviolet light from the sun is the main environmental factor. This genetic predisposition is starting to be understood, impacting not only on the risk of developing melanoma but also on the risk of developing other types of cancer, as well as on the prognosis of the disease.
Collapse
Affiliation(s)
- Sebastian Podlipnik
- Department of Dermatology, University of Barcelona, Hospital of Barcelona, Barcelona, Spain.,Unit of Melanoma, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Miriam Potrony
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain.,Department of Biochemistry and Molecular Genetics, Hospital of Barcelona, Barcelona, Spain
| | - Susana Puig
- Department of Dermatology, University of Barcelona, Hospital of Barcelona, Barcelona, Spain - .,Unit of Melanoma, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| |
Collapse
|
47
|
McKenna S, García-Gutiérrez L. Resistance to Targeted Therapy and RASSF1A Loss in Melanoma: What Are We Missing? Int J Mol Sci 2021; 22:5115. [PMID: 34066022 PMCID: PMC8150731 DOI: 10.3390/ijms22105115] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/26/2021] [Accepted: 05/06/2021] [Indexed: 12/20/2022] Open
Abstract
Melanoma is one of the most aggressive forms of skin cancer and is therapeutically challenging, considering its high mutation rate. Following the development of therapies to target BRAF, the most frequently found mutation in melanoma, promising therapeutic responses were observed. While mono- and combination therapies to target the MAPK cascade did induce a therapeutic response in BRAF-mutated melanomas, the development of resistance to MAPK-targeted therapies remains a challenge for a high proportion of patients. Resistance mechanisms are varied and can be categorised as intrinsic, acquired, and adaptive. RASSF1A is a tumour suppressor that plays an integral role in the maintenance of cellular homeostasis as a central signalling hub. RASSF1A tumour suppressor activity is commonly lost in melanoma, mainly by aberrant promoter hypermethylation. RASSF1A loss could be associated with several mechanisms of resistance to MAPK inhibition considering that most of the signalling pathways that RASSF1A controls are found to be altered targeted therapy resistant melanomas. Herein, we discuss resistance mechanisms in detail and the potential role for RASSF1A reactivation to re-sensitise BRAF mutant melanomas to therapy.
Collapse
Affiliation(s)
| | - Lucía García-Gutiérrez
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland;
| |
Collapse
|
48
|
Horn IP, Marks DL, Koenig AN, Hogenson TL, Almada LL, Goldstein LE, Romecin Duran PA, Vera R, Vrabel AM, Cui G, Rabe KG, Bamlet WR, Mer G, Sicotte H, Zhang C, Li H, Petersen GM, Fernandez-Zapico ME. A rare germline CDKN2A variant (47T>G; p16-L16R) predisposes carriers to pancreatic cancer by reducing cell cycle inhibition. J Biol Chem 2021; 296:100634. [PMID: 33823155 PMCID: PMC8121974 DOI: 10.1016/j.jbc.2021.100634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 03/29/2021] [Accepted: 04/01/2021] [Indexed: 11/30/2022] Open
Abstract
Germline mutations in CDKN2A, encoding the tumor suppressor p16, are responsible for a large proportion of familial melanoma cases and also increase risk of pancreatic cancer. We identified four families through pancreatic cancer probands that were affected by both cancers. These families bore a germline missense variant of CDKN2A (47T>G), encoding a p16-L16R mutant protein associated with high cancer occurrence. Here, we investigated the biological significance of this variant. When transfected into p16-null pancreatic cancer cells, p16-L16R was expressed at lower levels than wild-type (WT) p16. In addition, p16-L16R was unable to bind CDK4 or CDK6 compared with WT p16, as shown by coimmunoprecipitation assays and also was impaired in its ability to inhibit the cell cycle, as demonstrated by flow cytometry analyses. In silico molecular modeling predicted that the L16R mutation prevents normal protein folding, consistent with the observed reduction in expression/stability and diminished function of this mutant protein. We isolated normal dermal fibroblasts from members of the families expressing WT or L16R proteins to investigate the impact of endogenous p16-L16R mutant protein on cell growth. In culture, p16-L16R fibroblasts grew at a faster rate, and most survived until later passages than p16-WT fibroblasts. Further, western blotting demonstrated that p16 protein was detected at lower levels in p16-L16R than in p16-WT fibroblasts. Together, these results suggest that the presence of a CDKN2A (47T>G) mutant allele contributes to an increased risk of pancreatic cancer as a result of reduced p16 protein levels and diminished p16 tumor suppressor function.
Collapse
Affiliation(s)
- Isaac P Horn
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - David L Marks
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Amanda N Koenig
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Tara L Hogenson
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Luciana L Almada
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Lauren E Goldstein
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Paola A Romecin Duran
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Renzo Vera
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Anne M Vrabel
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Gaofeng Cui
- Division of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kari G Rabe
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - William R Bamlet
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Georges Mer
- Division of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Hugues Sicotte
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Cheng Zhang
- Division of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Hu Li
- Division of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Martin E Fernandez-Zapico
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
49
|
Sargen MR, Pfeiffer RM, Elder DE, Yang XR, Goldstein AM, Tucker MA. The Impact of Longitudinal Surveillance on Tumor Thickness for Melanoma-Prone Families with and without Pathogenic Germline Variants of CDKN2A and CDK4. Cancer Epidemiol Biomarkers Prev 2021; 30:676-681. [PMID: 33811164 DOI: 10.1158/1055-9965.epi-20-1521] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/18/2020] [Accepted: 01/28/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Skin cancer screening is routinely performed for members of melanoma-prone families, but longitudinal studies evaluating the efficacy of surveillance in this high-risk population are lacking. METHODS We evaluated thickness for first primary melanomas diagnosed in melanoma-prone families (≥2 individuals with melanoma) enrolled in NCT00040352 (NCI familial melanoma study) from 1976 through 2014; enrolled patients received routine skin cancer screening and education about skin self-exams. We used linear and ordinal logistic regression models adjusted for gender and age with a generalized estimating equations approach to report changes in thickness and tumor (T) stage over time, comparing outcomes for NCI cases diagnosed before (pre-study) versus after study participation (prospective) and for NCI cases versus nonfamilial cases [Surveillance, Epidemiology, and End Results (SEER) 9 registries]. RESULTS Tumor thickness was evaluated for 293 NCI (pre-study = 246; prospective = 47) patients. Compared with NCI pre-study cases, NCI prospective melanomas were thinner (0.6 vs. 1.1 mm; P < 0.001) and more likely to be T1 stage [39/47 (83%) vs. 98/246 (40%); P < 0.001]. Similar findings (P < 0.05) were observed for familial cases with and without germline CDKN2A and CDK4 mutations. Peters-Belson modeling suggested that calendar period effects of decreasing thickness in the general population (SEER 9) did not fully explain thickness trends in NCI families. CONCLUSIONS Participation in a longitudinal surveillance program providing skin cancer screening and education about skin self-exams was associated with thinner melanomas for members of melanoma-prone families. IMPACT The study findings support the clinical benefit of screening (physician and self) for this high-risk population.
Collapse
Affiliation(s)
- Michael R Sargen
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Rockville, Maryland.
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Rockville, Maryland
| | - David E Elder
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xiaohong R Yang
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Rockville, Maryland
| | - Alisa M Goldstein
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Rockville, Maryland
| | - Margaret A Tucker
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Rockville, Maryland
| |
Collapse
|
50
|
Chan SH, Chiang J, Ngeow J. CDKN2A germline alterations and the relevance of genotype-phenotype associations in cancer predisposition. Hered Cancer Clin Pract 2021; 19:21. [PMID: 33766116 PMCID: PMC7992806 DOI: 10.1186/s13053-021-00178-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/15/2021] [Indexed: 02/08/2023] Open
Abstract
Although CDKN2A is well-known as a susceptibility gene for melanoma and pancreatic cancer, germline variants have also been anecdotally associated with a broader range of neoplasms including neural system tumors, head and neck squamous cell carcinomas, breast carcinomas, as well as sarcomas. The CDKN2A gene encodes for two distinct tumor suppressor proteins, p16INK4A and p14ARF, however, the independent association of germline alterations affecting these two proteins with cancer is under-appreciated. Here, we reviewed CDKN2A germline alterations reported among individuals and families with cancer in the literature, specifically addressing the cancer phenotypes in relation to the molecular consequence on p16INK4A and p14ARF. While melanoma is observed to associate with variants affecting both p16INK4A and p14ARF transcripts, it is noted that variants affecting p14ARF are more frequently observed with a heterogenous range of cancers. Finally, we reflected on the implications of this inferred genotype-phenotype association in clinical practice and proposed that clinical management of CDKN2A germline variant carriers should involve dedicated cancer genetics services, with multidisciplinary input from various healthcare professionals.
Collapse
Affiliation(s)
- Sock Hoai Chan
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Jianbang Chiang
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Joanne Ngeow
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore.
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, 169857, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, 308232, Singapore.
| |
Collapse
|