Familial Cancer and Germline Genetics

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Cancer Biomarkers".

Deadline for manuscript submissions: closed (10 February 2022) | Viewed by 21329

Special Issue Editors


E-Mail Website
Guest Editor
1. Charles University Medical School in Pilsen, Pilsen, Czech Republic
2. Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
Interests: germline genetics; genetic predisposition; familial cancer; hereditary cancer; genome-wide association study; germline sequencing
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Co-Guest Editor
Hopp Children's Cancer Center (KiTZ), German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
Interests: germline genetics; whole genome sequencing; genome-wide association study; functional genomics

E-Mail Website
Co-Guest Editor
Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
Interests: cancer genetics and genomics

Special Issue Information

Dear Colleague, 

Both original articles and reviews are welcome. The focus of this Special Issue is two-fold. It will cover novel family studies on diverse cancers—novelty would be demonstrated through new types of familial clustering, high risk levels and statistical power, and possibly through populations not commonly reported. The technical requirements are diagnostic accuracy and unbiased selection of patients and family members. Results on germline genetics should be linked to families with either population, experimental, or in silico evidence on pathogenicity. Alternatively, large sequencing studies on defined sporadic cancers could be acceptable, particularly if justified through unique aspects such as rare cancers, whole-genome coverage, new populations, etc. Standard panel sequencing results are of lower priority.  

The guest editors will be happy to consider inquiries about article suitability (please address them to [email protected]).

Prof. Kari Hemminki
Dr. Asta Försti
Prof. Richard Houlston
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cancers is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • familial clustering
  • risk between different cancers
  • multiplex families
  • whole-genome sequencing
  • Mendelian cancers
  • high-risk variant
  • pathogenic variant

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

14 pages, 711 KiB  
Article
Familial Risks for Liver, Gallbladder and Bile Duct Cancers and for Their Risk Factors in Sweden, a Low-Incidence Country
by Kari Hemminki, Kristina Sundquist, Jan Sundquist, Asta Försti, Vaclav Liska, Akseli Hemminki and Xinjun Li
Cancers 2022, 14(8), 1938; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14081938 - 12 Apr 2022
Cited by 5 | Viewed by 1820
Abstract
We used the Swedish Cancer Registry data to address familial risks for concordant (same) and discordant (different) hepatobiliary cancers, including their associations with any other cancers and with known risk factors. Risks were also assessed between spouses. The analysis covered Swedish families and [...] Read more.
We used the Swedish Cancer Registry data to address familial risks for concordant (same) and discordant (different) hepatobiliary cancers, including their associations with any other cancers and with known risk factors. Risks were also assessed between spouses. The analysis covered Swedish families and their cancers between years 1958 and 2018. Adjusted familial risks were expressed as standardized incidence ratios (SIRs). Familial SIRs for concordant hepatocellular carcinoma (HCC) were 2.60, and for gallbladder cancer they were at the same level (2.76). Familial risk was also found for intrahepatic bile duct cancer and for female extrahepatic bile duct cancer. HCC was associated with lung and cervical cancers; extrahepatic bile duct and ampullary cancers were associated with colon and pancreatic cancers, suggesting Lynch syndrome. Among spouses, hepatobiliary cancer was associated with HCC, stomach, pancreatic, cervical and upper aerodigestive tract cancers. Among risk factors, family members diagnosed with alcohol-related disease showed association with HCC. The observed familial risks for hepatobiliary cancers were relatively high, and considering the poor prognosis of these cancers, prevention is of the utmost importance and should focus on moderation of alcohol consumption, vaccination/treatment of hepatitis viral infections and avoidance of overweight and other risk factors of type 2 diabetes. Full article
(This article belongs to the Special Issue Familial Cancer and Germline Genetics)
Show Figures

Figure 1

17 pages, 2984 KiB  
Article
Germline Variants of CYBA and TRPM4 Predispose to Familial Colorectal Cancer
by Lizhen Zhu, Beiping Miao, Dagmara Dymerska, Magdalena Kuswik, Elena Bueno-Martínez, Lara Sanoguera-Miralles, Eladio A. Velasco, Nagarajan Paramasivam, Matthias Schlesner, Abhishek Kumar, Ying Yuan, Jan Lubinski, Obul Reddy Bandapalli, Kari Hemminki and Asta Försti
Cancers 2022, 14(3), 670; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14030670 - 28 Jan 2022
Cited by 10 | Viewed by 3117
Abstract
Familial colorectal cancer (CRC) is only partially explained by known germline predisposing genes. We performed whole-genome sequencing in 15 Polish families of many affected individuals, without mutations in known CRC predisposing genes. We focused on loss-of-function variants and functionally characterized them. We identified [...] Read more.
Familial colorectal cancer (CRC) is only partially explained by known germline predisposing genes. We performed whole-genome sequencing in 15 Polish families of many affected individuals, without mutations in known CRC predisposing genes. We focused on loss-of-function variants and functionally characterized them. We identified a frameshift variant in the CYBA gene (c.246delC) in one family and a splice site variant in the TRPM4 gene (c.25–1 G > T) in another family. While both variants were absent or extremely rare in gene variant databases, we identified four additional Polish familial CRC cases and two healthy elderly individuals with the CYBA variant (odds ratio 2.46, 95% confidence interval 0.48–12.69). Both variants led to a premature stop codon and to a truncated protein. Functional characterization of the variants showed that knockdown of CYBA or TRPM4 depressed generation of reactive oxygen species (ROS) in LS174T and HT-29 cell lines. Knockdown of TRPM4 resulted in decreased MUC2 protein production. CYBA encodes a component in the NADPH oxidase system which generates ROS and controls, e.g., bacterial colonization in the gut. Germline CYBA variants are associated with early onset inflammatory bowel disease, supported with experimental evidence on loss of intestinal mucus barrier function due to ROS deficiency. TRPM4 encodes a calcium-activated ion channel, which, in a human colonic cancer cell line, controls calcium-mediated secretion of MUC2, a major component of intestinal mucus barrier. We suggest that the gene defects in CYBA and TRPM4 mechanistically involve intestinal barrier integrity through ROS and mucus biology, which converges in chronic bowel inflammation. Full article
(This article belongs to the Special Issue Familial Cancer and Germline Genetics)
Show Figures

Figure 1

13 pages, 949 KiB  
Article
Do BARD1 Mutations Confer an Elevated Risk of Prostate Cancer?
by Klaudia Stempa, Dominika Wokołorczyk, Wojciech Kluźniak, Emilia Rogoża-Janiszewska, Karolina Malińska, Helena Rudnicka, Tomasz Huzarski, Jacek Gronwald, Katarzyna Gliniewicz, Tadeusz Dębniak, Anna Jakubowska, Marcin Lener, Joanna Tomiczek-Szwiec, Paweł Domagała, Malwina Suszynska, Piotr Kozlowski, Tomasz Kluz, Mariusz Naczk, Jan Lubiński, Steven A. Narod, Mohammad R. Akbari, Cezary Cybulski and on behalf of the Polish Hereditary Prostate Cancer Consortiumadd Show full author list remove Hide full author list
Cancers 2021, 13(21), 5464; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13215464 - 30 Oct 2021
Cited by 1 | Viewed by 2097
Abstract
The current cancer testing gene panels tend to be comprehensive rather than site-specific. BARD1 is one of the genes commonly included in the multi-cancer testing panels. Mutations in BARD1 confer an increase in the risk for breast cancer, but it is not studied [...] Read more.
The current cancer testing gene panels tend to be comprehensive rather than site-specific. BARD1 is one of the genes commonly included in the multi-cancer testing panels. Mutations in BARD1 confer an increase in the risk for breast cancer, but it is not studied whether or not they predispose to prostate cancer. To establish if BARD1 mutations also predispose to prostate cancer, we screened BARD1 in 390 Polish patients with hereditary prostate cancer. No truncating mutations were identified by sequencing. We also genotyped 5715 men with unselected prostate cancer, and 10,252 controls for three recurrent BARD1 variants, including p.Q564X, p.R658C and p.R659=. Neither variant conferred elevated risk of prostate cancer (ORs between 0.84 and 1.15, p-values between 0.57 and 0.93) nor did they influence prostate cancer characteristics or survival. We conclude that men with a BARD1 mutation are not at elevated prostate cancer risk. It is not justified to inform men about increased prostate cancer risk in case of identification of a BARD1 mutation. However, a female relative of a man with a BARD1 mutation may benefit from this information and be tested for the mutation, because BARD1 is a breast cancer susceptibility gene. Full article
(This article belongs to the Special Issue Familial Cancer and Germline Genetics)
Show Figures

Figure 1

21 pages, 3313 KiB  
Article
Familial Risks and Proportions Describing Population Landscape of Familial Cancer
by Kari Hemminki, Kristina Sundquist, Jan Sundquist, Asta Försti, Akseli Hemminki and Xinjun Li
Cancers 2021, 13(17), 4385; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13174385 - 30 Aug 2021
Cited by 18 | Viewed by 2639
Abstract
Background: Familial cancer can be defined through the occurrence of the same cancer in two or more family members. We describe a nationwide landscape of familial cancer, including its frequency and the risk that it conveys, by using the largest family database in [...] Read more.
Background: Familial cancer can be defined through the occurrence of the same cancer in two or more family members. We describe a nationwide landscape of familial cancer, including its frequency and the risk that it conveys, by using the largest family database in the world with complete family structures and medically confirmed cancers. Patients/methods: We employed standardized incidence ratios (SIRs) to estimate familial risks for concordant cancer among first-degree relatives using the Swedish Cancer Registry from years 1958 through 2016. Results: Cancer risks in a 20–84 year old population conferred by affected parents or siblings were about two-fold compared to the risk for individuals with unaffected relatives. For small intestinal, testicular, thyroid and bone cancers and Hodgkin disease, risks were higher, five-to-eight-fold. Novel familial associations included adult bone, lip, pharyngeal, and connective tissue cancers. Familial cancers were found in 13.2% of families with cancer; for prostate cancer, the proportion was 26.4%. High-risk families accounted for 6.6% of all cancer families. Discussion/Conclusion: High-risk family history should be exceedingly considered for management, including targeted genetic testing. For the major proportion of familial clustering, where genetic testing may not be feasible, medical and behavioral intervention should be indicated for the patient and their family members, including screening recommendations and avoidance of carcinogenic exposure. Full article
(This article belongs to the Special Issue Familial Cancer and Germline Genetics)
Show Figures

Figure 1

12 pages, 613 KiB  
Article
Family History of Head and Neck Cancers
by Xinjun Li, Anni I. Koskinen, Otto Hemminki, Asta Försti, Jan Sundquist, Kristina Sundquist and Kari Hemminki
Cancers 2021, 13(16), 4115; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13164115 - 16 Aug 2021
Cited by 3 | Viewed by 2192
Abstract
Background: Head and neck cancers (HNCs) encompass a heterogeneous group of cancers between the mouth and larynx. Familial clustering in HNCs has been described, but how it influences individual sites and to which extent known risk factors, such as human papilloma virus (HPV) [...] Read more.
Background: Head and neck cancers (HNCs) encompass a heterogeneous group of cancers between the mouth and larynx. Familial clustering in HNCs has been described, but how it influences individual sites and to which extent known risk factors, such as human papilloma virus (HPV) infection, may contribute is not well established. Patients/methods: We employed standardized incidence ratios (SIRs) to estimate familial risks for HNC with same (concordant) and different (discordant) cancers among first-degree relatives using data from the Swedish Cancer Registry from 1958 to 2018. Results: Incidence for male and female oropharyngeal cancer increased close to four-fold in the past 39 years. Familial HNC was found in 3.4% of the study population, with an overall familial SIR of 1.78. Patients with concordant nasopharyngeal cancer showed a high risk of 23.97, followed by hypopharyngeal cancer (5.43). The husbands of wives with cervical cancer had an increased risk of oropharyngeal cancer. Discussion/Conclusion: Nasopharyngeal cancers lacked associations with lifestyle or HPV associated cancers, suggesting a role for germline genetics, which was also true for the high-risk families of three HNC patients. In the Swedish population with low smoking levels, HPV is becoming a dominant risk factor, emphasizing the need for sexual hygiene and HPV vaccination. Full article
(This article belongs to the Special Issue Familial Cancer and Germline Genetics)
Show Figures

Figure 1

12 pages, 1172 KiB  
Article
Familial Risk and Heritability of Hematologic Malignancies in the Nordic Twin Study of Cancer
by Signe B. Clemmensen, Jennifer R. Harris, Jonas Mengel-From, Wagner H. Bonat, Henrik Frederiksen, Jaakko Kaprio and Jacob v. B. Hjelmborg
Cancers 2021, 13(12), 3023; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13123023 - 16 Jun 2021
Cited by 3 | Viewed by 3338
Abstract
We aimed to explore the genetic and environmental contributions to variation in the risk of hematologic malignancies and characterize familial dependence within and across hematologic malignancies. The study base included 316,397 individual twins from the Nordic Twin Study of Cancer with a median [...] Read more.
We aimed to explore the genetic and environmental contributions to variation in the risk of hematologic malignancies and characterize familial dependence within and across hematologic malignancies. The study base included 316,397 individual twins from the Nordic Twin Study of Cancer with a median of 41 years of follow-up: 88,618 (28%) of the twins were monozygotic, and 3459 hematologic malignancies were reported. We estimated the cumulative incidence by age, familial risk, and genetic and environmental variance components of hematologic malignancies accounting for competing risk of death. The lifetime risk of any hematologic malignancy was 2.5% (95% CI 2.4–2.6%), as in the background population. This risk was elevated to 4.5% (95% CI 3.1–6.5%) conditional on hematologic malignancy in a dizygotic co-twin and was even greater at 7.6% (95% CI 4.8–11.8%) if a monozygotic co-twin had a hematologic malignancy. Heritability of the liability to develop any hematologic malignancy was 24% (95% CI 14–33%). This estimate decreased across age, from approximately 55% at age 40 to about 20–25% after age 55, when it seems to stabilize. In this largest ever studied twin cohort with the longest follow-up, we found evidence for familial risk of hematologic malignancies. The discovery of decreasing familial predisposition with increasing age underscores the importance of cancer surveillance in families with hematological malignancies. Full article
(This article belongs to the Special Issue Familial Cancer and Germline Genetics)
Show Figures

Figure 1

12 pages, 814 KiB  
Article
Rare Germline Variants in Chordoma-Related Genes and Chordoma Susceptibility
by Sally Yepes, Nirav N. Shah, Jiwei Bai, Hela Koka, Chuzhong Li, Songbai Gui, Mary Lou McMaster, Yanzi Xiao, Kristine Jones, Mingyi Wang, Aurelie Vogt, Bin Zhu, Bin Zhu, Amy Hutchinson, Meredith Yeager, Belynda Hicks, Brian Carter, Neal D. Freedman, Laura Beane-Freeman, Stephen J. Chanock, Yazhuo Zhang, Dilys M. Parry, Xiaohong R. Yang and Alisa M. Goldsteinadd Show full author list remove Hide full author list
Cancers 2021, 13(11), 2704; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13112704 - 30 May 2021
Cited by 8 | Viewed by 2952
Abstract
Background: Chordoma is a rare bone cancer with an unknown etiology. TBXT is the only chordoma susceptibility gene identified to date; germline single nucleotide variants and copy number variants in TBXT have been associated with chordoma susceptibility in familial and sporadic chordoma. However, [...] Read more.
Background: Chordoma is a rare bone cancer with an unknown etiology. TBXT is the only chordoma susceptibility gene identified to date; germline single nucleotide variants and copy number variants in TBXT have been associated with chordoma susceptibility in familial and sporadic chordoma. However, the genetic susceptibility of chordoma remains largely unknown. In this study, we investigated rare germline genetic variants in genes involved in TBXT/chordoma-related signaling pathways and other biological processes in chordoma patients from North America and China. Methods: We identified variants that were very rare in general population and internal control datasets and showed evidence for pathogenicity in 265 genes in a whole exome sequencing (WES) dataset of 138 chordoma patients of European ancestry and in a whole genome sequencing (WGS) dataset of 80 Chinese patients with skull base chordoma. Results: Rare and likely pathogenic variants were identified in 32 of 138 European ancestry patients (23%), including genes that are part of notochord development, PI3K/AKT/mTOR, Sonic Hedgehog, SWI/SNF complex and mesoderm development pathways. Rare pathogenic variants in COL2A1, EXT1, PDK1, LRP2, TBXT and TSC2, among others, were also observed in Chinese patients. Conclusion: We identified several rare loss-of-function and predicted deleterious missense variants in germline DNA from patients with chordoma, which may influence chordoma predisposition and reflect a complex susceptibility, warranting further investigation in large studies. Full article
(This article belongs to the Special Issue Familial Cancer and Germline Genetics)
Show Figures

Figure 1

11 pages, 1663 KiB  
Article
A Rare Variant in ERF (rs144812092) Predisposes to Prostate and Bladder Cancers in an Extended Pedigree
by Lisa Anne Cannon-Albright, Craig Carl Teerlink, Jeff Stevens, Franklin W. Huang, Csilla Sipeky, Johanna Schleutker, Rolando Hernandez, Julio Facelli, Neeraj Agarwal and Donald L. Trump
Cancers 2021, 13(10), 2399; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13102399 - 15 May 2021
Cited by 4 | Viewed by 1815
Abstract
Pairs of related bladder cancer cases who belong to pedigrees with an excess of bladder cancer were sequenced to identify rare, shared variants as candidate predisposition variants. Candidate variants were tested for association with bladder cancer risk. A validated variant was assayed for [...] Read more.
Pairs of related bladder cancer cases who belong to pedigrees with an excess of bladder cancer were sequenced to identify rare, shared variants as candidate predisposition variants. Candidate variants were tested for association with bladder cancer risk. A validated variant was assayed for segregation to other related cancer cases, and the predicted protein structure of this variant was analyzed. This study of affected bladder cancer relative pairs from high-risk pedigrees identified 152 bladder cancer predisposition candidate variants. One variant in ERF (ETS Repressing Factor) was significantly associated with bladder cancer risk in an independent population, was observed to segregate with bladder and prostate cancer in relatives, and showed evidence for altering the function of the associated protein. This finding of a rare variant in ERF that is strongly associated with bladder and prostate cancer risk in an extended pedigree both validates ERF as a cancer predisposition gene and shows the continuing value of analyzing affected members of high-risk pedigrees to identify and validate rare cancer predisposition variants. Full article
(This article belongs to the Special Issue Familial Cancer and Germline Genetics)
Show Figures

Figure 1

Back to TopTop