101
|
Climente-González H, Lonjou C, Lesueur F, Stoppa-Lyonnet D, Andrieu N, Azencott CA. Boosting GWAS using biological networks: A study on susceptibility to familial breast cancer. PLoS Comput Biol 2021; 17:e1008819. [PMID: 33735170 PMCID: PMC8009366 DOI: 10.1371/journal.pcbi.1008819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 03/30/2021] [Accepted: 02/18/2021] [Indexed: 11/20/2022] Open
Abstract
Genome-wide association studies (GWAS) explore the genetic causes of complex diseases. However, classical approaches ignore the biological context of the genetic variants and genes under study. To address this shortcoming, one can use biological networks, which model functional relationships, to search for functionally related susceptibility loci. Many such network methods exist, each arising from different mathematical frameworks, pre-processing steps, and assumptions about the network properties of the susceptibility mechanism. Unsurprisingly, this results in disparate solutions. To explore how to exploit these heterogeneous approaches, we selected six network methods and applied them to GENESIS, a nationwide French study on familial breast cancer. First, we verified that network methods recovered more interpretable results than a standard GWAS. We addressed the heterogeneity of their solutions by studying their overlap, computing what we called the consensus. The key gene in this consensus solution was COPS5, a gene related to multiple cancer hallmarks. Another issue we observed was that network methods were unstable, selecting very different genes on different subsamples of GENESIS. Therefore, we proposed a stable consensus solution formed by the 68 genes most consistently selected across multiple subsamples. This solution was also enriched in genes known to be associated with breast cancer susceptibility (BLM, CASP8, CASP10, DNAJC1, FGFR2, MRPS30, and SLC4A7, P-value = 3 × 10-4). The most connected gene was CUL3, a regulator of several genes linked to cancer progression. Lastly, we evaluated the biases of each method and the impact of their parameters on the outcome. In general, network methods preferred highly connected genes, even after random rewirings that stripped the connections of any biological meaning. In conclusion, we present the advantages of network-guided GWAS, characterize their shortcomings, and provide strategies to address them. To compute the consensus networks, implementations of all six methods are available at https://github.com/hclimente/gwas-tools.
Collapse
Affiliation(s)
- Héctor Climente-González
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
- RIKEN Center for Advanced Intelligence Project (AIP), Tokyo, Japan
| | - Christine Lonjou
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Fabienne Lesueur
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | | | - Dominique Stoppa-Lyonnet
- Service de Génétique, Institut Curie, Paris, France
- INSERM, U830, Paris, France
- Université Paris Descartes, Paris, France
| | - Nadine Andrieu
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Chloé-Agathe Azencott
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| |
Collapse
|
102
|
Shin W, Jeong G, Son Y, Seo SS, Kang S, Park SY, Lim MC. The Knowledge and Attitude of Patients Diagnosed with Epithelial Ovarian Cancer towards Genetic Testing. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18052312. [PMID: 33652933 PMCID: PMC7956717 DOI: 10.3390/ijerph18052312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 11/16/2022]
Abstract
This study assessed the knowledge and attitude of patients with ovarian cancer (OC) toward OC and next generation sequencing (NGS). The data, including characteristics of patients, their knowledge about OC and their knowledge and attitude of NGS, were collected from June to October 2018. Of the 103 participants, 70.9% (n = 73) had cancer within the second-degree relatives, and 18.4% (n = 19) had BRCA pathogenic mutations. The percentage of right answer for the knowledge about OC and NGS was 64.7% (11/17) and 50% (6/12), respectively. The median number of patients who had positive expectations for the genetic test was 34 (range, 22-44). Based on a first-degree familial history, patients had a different degree of knowledge about OC (11 vs. 8.5, p = 0.026) and NGS (6.5 vs. 5, p = 0.011), but patients with a BRCA pathogenic mutation did not have a different degree of knowledge about OC and NGS panel testing. High-income families had a more positive attitude towards the genetic test than low-income families (p = 0.005). Women with OC do not have enough knowledge about OC (11/17, 64.7%) and NGS (6/12, 50%) but they showed a positive attitude toward the NGS test. These women need OC and NGS educational intervention.
Collapse
Affiliation(s)
- Wonkyo Shin
- Center for Gynecologic Cancer, National Cancer Center, 323 Ilsan-ro, Goyang 10408, Korea; (W.S.); (G.J.); (S.-S.S.); (S.K.); (S.-Y.P.)
| | - Gowoon Jeong
- Center for Gynecologic Cancer, National Cancer Center, 323 Ilsan-ro, Goyang 10408, Korea; (W.S.); (G.J.); (S.-S.S.); (S.K.); (S.-Y.P.)
| | - Yedong Son
- College of Nursing, Woosuk University, Wanju 55338, Korea;
| | - Sang-Soo Seo
- Center for Gynecologic Cancer, National Cancer Center, 323 Ilsan-ro, Goyang 10408, Korea; (W.S.); (G.J.); (S.-S.S.); (S.K.); (S.-Y.P.)
| | - Sokbom Kang
- Center for Gynecologic Cancer, National Cancer Center, 323 Ilsan-ro, Goyang 10408, Korea; (W.S.); (G.J.); (S.-S.S.); (S.K.); (S.-Y.P.)
- Division of Precision Medicine, Research Institute, National Cancer Center, 323 Ilsan-ro, Goyang 10408, Korea
- Department of Cancer Control & Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Korea
| | - Sang-Yoon Park
- Center for Gynecologic Cancer, National Cancer Center, 323 Ilsan-ro, Goyang 10408, Korea; (W.S.); (G.J.); (S.-S.S.); (S.K.); (S.-Y.P.)
| | - Myong Cheol Lim
- Center for Gynecologic Cancer, National Cancer Center, 323 Ilsan-ro, Goyang 10408, Korea; (W.S.); (G.J.); (S.-S.S.); (S.K.); (S.-Y.P.)
- Department of Cancer Control & Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Korea
- Center for Clinical Trials, Hospital, National Cancer Center, 323 Ilsan-ro, Goyang 10408, Korea
- Division of Tumor Immunology, National Cancer Center, 323 Ilsan-ro, Goyang 10408, Korea
- Correspondence: ; Tel.: +82-31-920-1763
| |
Collapse
|
103
|
De Angelis C, Nardelli C, Concolino P, Pagliuca M, Setaro M, De Paolis E, De Placido P, Forestieri V, Scaglione GL, Ranieri A, Lombardo B, Pastore L, De Placido S, Capoluongo E. Case Report: Detection of a Novel Germline PALB2 Deletion in a Young Woman With Hereditary Breast Cancer: When the Patient's Phenotype History Doesn't Lie. Front Oncol 2021; 11:602523. [PMID: 33718150 PMCID: PMC7943848 DOI: 10.3389/fonc.2021.602523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 02/05/2021] [Indexed: 11/23/2022] Open
Abstract
The partner and localizer of BRCA2 (PALB2) is a major BRCA2 binding partner that participates in homologous recombination repair in response to DNA double-strand breaks. Germline alterations of the PALB2 gene have recently been associated with a high risk of developing breast cancer. We investigated a 37-year-old Caucasian woman with breast cancer and family history of breast cancer using targeted next generation sequencing. A novel heterozygous deletion involving exons 5 and 6 was found in the PALB2 gene, and resulted in the production of a truncated PALB2 protein. These findings expand the mutational spectra of PALB2-associated breast cancer, and may improve the mutation-based screening and genetic diagnosis of breast cancer.
Collapse
Affiliation(s)
- Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Carmela Nardelli
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Paola Concolino
- Molecular and Genomic Diagnostics Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Martina Pagliuca
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | - Elisa De Paolis
- Molecular and Genomic Diagnostics Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Pietro De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Valeria Forestieri
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | - Annalisa Ranieri
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate, Naples, Italy
| | - Barbara Lombardo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate, Naples, Italy
| | - Lucio Pastore
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate, Naples, Italy
| | - Sabino De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Ettore Capoluongo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate, Naples, Italy
| |
Collapse
|
104
|
Li W, Gu X, Liu C, Shi Y, Wang P, Zhang N, Wu R, Leng L, Xie B, Song C, Li M. A synergetic effect of BARD1 mutations on tumorigenesis. Nat Commun 2021; 12:1243. [PMID: 33623049 PMCID: PMC7902612 DOI: 10.1038/s41467-021-21519-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 01/28/2021] [Indexed: 12/19/2022] Open
Abstract
To date, a large number of mutations have been screened from breast and ovarian cancer patients. However, most of them are classified into benign or unidentified alterations due to their undetectable phenotypes. Whether and how they could cause tumors remains unknown, and this significantly limits diagnosis and therapy. Here, in a study of a family with hereditary breast and ovarian cancer, we find that two BARD1 mutations, P24S and R378S, simultaneously exist in cis in surviving cancer patients. Neither of the single mutations causes a functional change, but together they synergetically impair the DNA damage response and lead to tumors in vitro and in vivo. Thus, our report not only demonstrates that BARD1 defects account for tumorigenesis but also uncovers the potential risk of synergetic effects between the large number of cis mutations in individual genes in the human genome.
Collapse
Affiliation(s)
- Wenjing Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, China
| | - Xiaoyang Gu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, China
| | - Chunhong Liu
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yanyan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | - Pan Wang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Na Zhang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Rui Wu
- Department of Pathology, Peking University Third Hospital, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Liang Leng
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China
| | - Bingteng Xie
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Chen Song
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Mo Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, China.
| |
Collapse
|
105
|
Tomasova K, Kroupa M, Forsti A, Vodicka P, Vodickova L. Telomere maintenance in interplay with DNA repair in pathogenesis and treatment of colorectal cancer. Mutagenesis 2021; 35:261-271. [PMID: 32083302 DOI: 10.1093/mutage/geaa005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/29/2020] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) continues to be one of the leading malignancies and causes of tumour-related deaths worldwide. Both impaired DNA repair mechanisms and disrupted telomere length homeostasis represent key culprits in CRC initiation, progression and prognosis. Mechanistically, altered DNA repair results in the accumulation of mutations in the genome and, ultimately, in genomic instability. DNA repair also determines the response to chemotherapeutics in CRC treatment, suggesting its utilisation in the prediction of therapy response and individual approach to patients. Telomere attrition resulting in replicative senescence, simultaneously by-passing cell cycle checkpoints, is a hallmark of malignant transformation of the cell. Telomerase is almost ubiquitous in advanced solid cancers, including CRC, and its expression is fundamental to cell immortalisation. Therefore, there is a persistent effort to develop therapeutics, which are telomerase-specific and gentle to non-malignant tissues. However, in practice, we are still at the level of clinical trials. The current state of knowledge and the route, which the research takes, gives us a positive perspective that the problem of molecular models of telomerase activation and telomere length stabilisation will finally be solved. We summarise the current literature herein, by pointing out the crosstalk between proteins involved in DNA repair and telomere length homeostasis in relation to CRC.
Collapse
Affiliation(s)
- Kristyna Tomasova
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine, The Czech Academy of Sciences, Vídeňská, Praha, Czech Republic.,Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Alej Svobody, Plzeň, Czech Republic
| | - Michal Kroupa
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine, The Czech Academy of Sciences, Vídeňská, Praha, Czech Republic.,Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Alej Svobody, Plzeň, Czech Republic
| | - Asta Forsti
- Hopp Children's Cancer Center (KiTZ), Im Neuenheimer Feld, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Im Neuenheimer Feld, Heidelberg, Germany
| | - Pavel Vodicka
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine, The Czech Academy of Sciences, Vídeňská, Praha, Czech Republic.,Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Alej Svobody, Plzeň, Czech Republic.,Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov, Praha, Czech Republic
| | - Ludmila Vodickova
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine, The Czech Academy of Sciences, Vídeňská, Praha, Czech Republic.,Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Alej Svobody, Plzeň, Czech Republic.,Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov, Praha, Czech Republic
| |
Collapse
|
106
|
Kato S. Tumour-Agnostic Therapy for Pancreatic Cancer and Biliary Tract Cancer. Diagnostics (Basel) 2021; 11:252. [PMID: 33562094 PMCID: PMC7914923 DOI: 10.3390/diagnostics11020252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/31/2021] [Accepted: 02/04/2021] [Indexed: 12/15/2022] Open
Abstract
The prognosis of patients with solid tumours has remarkably improved with the development of molecular-targeted drugs and immune checkpoint inhibitors. However, the improvements in the prognosis of pancreatic cancer and biliary tract cancer is delayed compared to other carcinomas, and the 5-year survival rates of distal-stage disease are approximately 10 and 20%, respectively. However, a comprehensive analysis of tumour cells using The Cancer Genome Atlas (TCGA) project has led to the identification of various driver mutations. Evidently, few mutations exist across organs, and basket trials targeting driver mutations regardless of the primary organ are being actively conducted. Such basket trials not only focus on the gate keeper-type oncogene mutations, such as HER2 and BRAF, but also focus on the caretaker-type tumour suppressor genes, such as BRCA1/2, mismatch repair-related genes, which cause hereditary cancer syndrome. As oncogene panel testing is a vital approach in routine practice, clinicians should devise a strategy for improved understanding of the cancer genome. Here, the gene mutation profiles of pancreatic cancer and biliary tract cancer have been outlined and the current status of tumour-agnostic therapy in these cancers has been reported.
Collapse
Affiliation(s)
- Shunsuke Kato
- Department of Clinical Oncology, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
107
|
Miklikova S, Trnkova L, Plava J, Bohac M, Kuniakova M, Cihova M. The Role of BRCA1/2-Mutated Tumor Microenvironment in Breast Cancer. Cancers (Basel) 2021; 13:575. [PMID: 33540843 PMCID: PMC7867315 DOI: 10.3390/cancers13030575] [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: 12/31/2020] [Revised: 01/23/2021] [Accepted: 01/29/2021] [Indexed: 12/15/2022] Open
Abstract
Taking into account the factors of high incidence rate, prevalence and mortality, breast cancer represents a crucial social and economic burden. Most cases of breast cancer develop as a consequence of somatic mutations accumulating in mammary epithelial cells throughout lifetime and approximately 5-10% can be ascribed to monogenic predispositions. Even though the role of genetic predispositions in breast cancer is well described in the context of genetics, very little is known about the role of the microenvironment carrying the same aberrant cells impaired by the germline mutation in the breast cancer development and progression. Based on the clinical observations, carcinomas carrying mutations in hereditary tumor-suppressor genes involved in maintaining genome integrity such as BRCA1/2 have worse prognosis and aggressive behavior. One of the mechanisms clarifying the aggressive nature of BRCA-associated tumors implies alterations within the surrounding adipose tissue itself. The objective of this review is to look at the role of BRCA1/2 mutations in the context of breast tumor microenvironment and plausible mechanisms by which it contributes to the aggressive behavior of the tumor cells.
Collapse
Affiliation(s)
- Svetlana Miklikova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, 84505 Bratislava, Slovakia; (S.M.); (L.T.); (J.P.)
| | - Lenka Trnkova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, 84505 Bratislava, Slovakia; (S.M.); (L.T.); (J.P.)
| | - Jana Plava
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, 84505 Bratislava, Slovakia; (S.M.); (L.T.); (J.P.)
| | - Martin Bohac
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, National Cancer Institute, Klenova 1, 83310 Bratislava, Slovakia;
- Department of Oncosurgery, National Cancer Institute, Klenova 1, 83310 Bratislava, Slovakia
- Regenmed Ltd., Medena 29, 81108 Bratislava, Slovakia
| | - Marcela Kuniakova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 81108 Bratislava, Slovakia;
| | - Marina Cihova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, 84505 Bratislava, Slovakia; (S.M.); (L.T.); (J.P.)
| |
Collapse
|
108
|
Zarrizi R, Higgs MR, Voßgröne K, Rossing M, Bertelsen B, Bose M, Kousholt AN, Rösner H, Network TC, Ejlertsen B, Stewart GS, Nielsen FC, Sørensen CS. Germline RBBP8 variants associated with early-onset breast cancer compromise replication fork stability. J Clin Invest 2021; 130:4069-4080. [PMID: 32379725 DOI: 10.1172/jci127521] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Haploinsufficiency of factors governing genome stability underlies hereditary breast and ovarian cancer. One significant pathway that is disabled as a result is homologous recombination repair (HRR). With the aim of identifying new candidate genes, we examined early-onset breast cancer patients negative for BRCA1 and BRCA2 pathogenic variants. Here, we focused on CtIP (RBBP8 gene), which mediates HRR through the end resection of DNA double-strand breaks (DSBs). Notably, these patients exhibited a number of rare germline RBBP8 variants. Functional analysis revealed that these variants did not affect DNA DSB end resection efficiency. However, expression of a subset of variants led to deleterious nucleolytic degradation of stalled DNA replication forks in a manner similar to that of cells lacking BRCA1 or BRCA2. In contrast to BRCA1 and BRCA2, CtIP deficiency promoted the helicase-driven destabilization of RAD51 nucleofilaments at damaged DNA replication forks. Taken together, our work identifies CtIP as a critical regulator of DNA replication fork integrity, which, when compromised, may predispose to the development of early-onset breast cancer.
Collapse
Affiliation(s)
- Reihaneh Zarrizi
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Martin R Higgs
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Karolin Voßgröne
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Maria Rossing
- Centre for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Birgitte Bertelsen
- Centre for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Muthiah Bose
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | | | - Heike Rösner
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | | | - Bent Ejlertsen
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Grant S Stewart
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Finn Cilius Nielsen
- Centre for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Claus S Sørensen
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
109
|
Chen Z, Wang X, Li X, Zhou Y, Chen K. Deep exploration of PARP inhibitors in breast cancer: monotherapy and combination therapy. J Int Med Res 2021; 49:300060521991019. [PMID: 33541181 PMCID: PMC8164563 DOI: 10.1177/0300060521991019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 01/05/2021] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Nearly 5% of patients with breast cancer carry germline BRCA mutations, which are more common in triple-negative breast cancer (TNBC). Previous clinical trials demonstrated the therapeutic efficacy of poly (ADP-ribose) polymerase inhibitors (PARPis) against BRCA-mutated metastatic breast cancer. The current study conducted a systemic review and meta-analysis of the clinical efficiency and safety of PARPis, either alone or combined with chemotherapy, in patients with TNBC. METHODS We searched PubMed, EMBASE, and ClinicalTrials.gov to identify randomized controlled trials comparing PARPi therapy with chemotherapy, and comparisons of chemotherapy plus PARPis with chemotherapy alone were included. The study endpoints included the clinical response, progression-free survival, and adverse event rates. RESULTS PARPi therapy was revealed to improve progression-free survival in patients with advanced breast cancer, either alone or in combination with chemotherapy. Subgroup analysis illustrated that patients with mutant BRCA1 and mutant BRCA2 and those who had not been treated with platinum-based agents could specifically benefit from PARPis. CONCLUSION PARPi monotherapy can significantly improve clinical outcomes in patients with advanced breast cancer, especially those with TNBC, those who had not previously received platinum therapy, and those with mutant BRCA1/2. PARPis combined with chemotherapy represent new treatment options for patients with advanced cancer.
Collapse
Affiliation(s)
- Zheling Chen
- Department of Medical Oncology, Zhejiang Provincial People’s
Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang,
People’s Republic of China
| | - Xiao Wang
- Department of Medical Oncology, Zhejiang Provincial People’s
Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang,
People’s Republic of China
| | - Xiao Li
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang
Provincial People's Hospital, People's Hospital of Hangzhou Medical College,
Hangzhou, Zhejiang, People’s Republic of China
| | - Yucheng Zhou
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang
Provincial People's Hospital, People's Hospital of Hangzhou Medical College,
Hangzhou, Zhejiang, People’s Republic of China
| | - Ke Chen
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang
Provincial People's Hospital, People's Hospital of Hangzhou Medical College,
Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
110
|
Heinze K, Rengsberger M, Gajda M, Jansen L, Osmers L, Oliveira-Ferrer L, Schmalfeldt B, Dürst M, Häfner N, Runnebaum IB. CAMK2N1/RUNX3 methylation is an independent prognostic biomarker for progression-free and overall survival of platinum-sensitive epithelial ovarian cancer patients. Clin Epigenetics 2021; 13:15. [PMID: 33482905 PMCID: PMC7824928 DOI: 10.1186/s13148-021-01006-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND To date, no predictive or prognostic molecular biomarkers except BRCA mutations are clinically established for epithelial ovarian cancer (EOC) despite being the deadliest gynecological malignancy. Aim of this biomarker study was the analysis of DNA methylation biomarkers for their prognostic value independent from clinical variables in a heterogeneous cohort of 203 EOC patients from two university medical centers. RESULTS The marker combination CAMK2N1/RUNX3 exhibited a significant prognostic value for progression-free (PFS) and overall survival (OS) of sporadic platinum-sensitive EOC (n = 188) both in univariate Kaplan-Meier (LogRank p < 0.05) and multivariate Cox regression analysis (p < 0.05; hazard ratio HR = 1.587). KRT86 methylation showed a prognostic value only in univariate analysis because of an association with FIGO staging (Fisher's exact test p < 0.01). Thus, it may represent a marker for EOC staging. Dichotomous prognostic values were observed for KATNAL2 methylation depending on BRCA aberrations. KATNAL2 methylation exhibited a negative prognostic value for PFS in sporadic EOC patients without BRCA1 methylation (HR 1.591, p = 0.012) but positive prognostic value in sporadic EOC with BRCA1 methylation (HR 0.332, p = 0.04) or BRCA-mutated EOC (HR 0.620, n.s.). CONCLUSION The retrospective analysis of 188 sporadic platinum-sensitive EOC proved an independent prognostic value of the methylation marker combination CAMK2N1/RUNX3 for PFS and OS. If validated prospectively this combination may identify EOC patients with worse prognosis after standard therapy potentially benefiting from intensive follow-up, maintenance therapies or inclusion in therapeutic studies. The dichotomous prognostic value of KATNAL2 should be validated in larger sample sets of EOC.
Collapse
Affiliation(s)
- Karolin Heinze
- Department of Gynecology and Reproduction Medicine, Jena University Hospital-Friedrich Schiller University Jena, 07747, Jena, Germany
| | - Matthias Rengsberger
- Department of Gynecology and Reproduction Medicine, Jena University Hospital-Friedrich Schiller University Jena, 07747, Jena, Germany
| | - Mieczyslaw Gajda
- Department of Forensic Medicine, Section of Pathology, Jena University Hospital - Friedrich Schiller University Jena, 07747, Jena, Germany
| | - Lars Jansen
- Department of Gynecology and Reproduction Medicine, Jena University Hospital-Friedrich Schiller University Jena, 07747, Jena, Germany
| | - Linea Osmers
- Department of Gynecology and Reproduction Medicine, Jena University Hospital-Friedrich Schiller University Jena, 07747, Jena, Germany
| | - Leticia Oliveira-Ferrer
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Barbara Schmalfeldt
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Matthias Dürst
- Department of Gynecology and Reproduction Medicine, Jena University Hospital-Friedrich Schiller University Jena, 07747, Jena, Germany
| | - Norman Häfner
- Department of Gynecology and Reproduction Medicine, Jena University Hospital-Friedrich Schiller University Jena, 07747, Jena, Germany.
| | - Ingo B Runnebaum
- Department of Gynecology and Reproduction Medicine, Jena University Hospital-Friedrich Schiller University Jena, 07747, Jena, Germany.
| |
Collapse
|
111
|
Incorvaia L, Fanale D, Bono M, Calò V, Fiorino A, Brando C, Corsini LR, Cutaia S, Cancelliere D, Pivetti A, Filorizzo C, La Mantia M, Barraco N, Cusenza S, Badalamenti G, Russo A, Bazan V. BRCA1/2 pathogenic variants in triple-negative versus luminal-like breast cancers: genotype-phenotype correlation in a cohort of 531 patients. Ther Adv Med Oncol 2020; 12:1758835920975326. [PMID: 33403015 PMCID: PMC7747114 DOI: 10.1177/1758835920975326] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/29/2020] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Several available data suggest the association between specific molecular subtypes and BRCA1/2 mutational status. Previous investigations showed the association between BRCA1/2 pathogenic variants (PVs) in specific genomic regions and phenotypic variations of cancer relative risk, while the role of PV type and location in determining the breast cancer (BC) phenotypic features remains still unclear. The aim of this research was to describe the germline BRCA1/2 PVs in triple-negative breast cancer (TNBC) versus luminal-like BC and their potential leverage on BC phenotype. PATIENTS & METHODS We retrospectively collected and analyzed all clinical information of 531 patients with BC genetically tested for germline BRCA1/2 PVs by Next-Generation Sequencing analysis at University Hospital Policlinico "P. Giaccone" of Palermo (Sicily) from January 2016 to February 2020. RESULTS Our results corroborate the evidence that BRCA1-related tumors often have a profile which resembles the TNBC subtype, whereas BRCA2-associated tumors have a profile that resembles luminal-like BC, especially the Luminal B subtype. Interestingly, our findings suggest that the PVs identified in TNBC were not largely overlapping with those in luminal-like tumors. Differences in the frequency of two PVs potentially associated with different molecular tumor subtypes were observed. BRCA1-633delC was detected with relatively higher prevalence in patients with TNBC, whereas BRCA2-1466delT was found mainly in Luminal B tumors, but in no TNBC patient. CONCLUSION Future studies examining the type and location of BRCA1/2 PVs within different molecular subtypes are required to verify our hypothesis and could provide an interesting insight into the complex topic of genotype-phenotype correlations. Additionally, a more in-depth understanding of the potential correlations between BRCA PVs and clinical and phenotypic features of hereditary BC syndrome patients could be the key to develop better strategies of prevention and surveillance in BRCA-positive carriers without disease.
Collapse
Affiliation(s)
- Lorena Incorvaia
- Section of Medical Oncology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Daniele Fanale
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Marco Bono
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Valentina Calò
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Alessia Fiorino
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Chiara Brando
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Lidia Rita Corsini
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Sofia Cutaia
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Daniela Cancelliere
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Alessia Pivetti
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Clarissa Filorizzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Maria La Mantia
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Nadia Barraco
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Stefania Cusenza
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| | - Viviana Bazan
- Section of Medical Oncology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy
| |
Collapse
|
112
|
Sanoguera-Miralles L, Valenzuela-Palomo A, Bueno-Martínez E, Llovet P, Díez-Gómez B, Caloca MJ, Pérez-Segura P, Fraile-Bethencourt E, Colmena M, Carvalho S, Allen J, Easton DF, Devilee P, Vreeswijk MPG, de la Hoya M, Velasco EA. Comprehensive Functional Characterization and Clinical Interpretation of 20 Splice-Site Variants of the RAD51C Gene. Cancers (Basel) 2020; 12:E3771. [PMID: 33333735 PMCID: PMC7765170 DOI: 10.3390/cancers12123771] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
Hereditary breast and/or ovarian cancer is a highly heterogeneous disease with more than 10 known disease-associated genes. In the framework of the BRIDGES project (Breast Cancer Risk after Diagnostic Gene Sequencing), the RAD51C gene has been sequenced in 60,466 breast cancer patients and 53,461 controls. We aimed at functionally characterizing all the identified genetic variants that are predicted to disrupt the splicing process. Forty RAD51C variants of the intron-exon boundaries were bioinformatically analyzed, 20 of which were selected for splicing functional assays. To test them, a splicing reporter minigene with exons 2 to 8 was designed and constructed. This minigene generated a full-length transcript of the expected size (1062 nucleotides), sequence, and structure (Vector exon V1- RAD51C exons_2-8- Vector exon V2). The 20 candidate variants were genetically engineered into the wild type minigene and functionally assayed in MCF-7 cells. Nineteen variants (95%) impaired splicing, while 18 of them produced severe splicing anomalies. At least 35 transcripts were generated by the mutant minigenes: 16 protein-truncating, 6 in-frame, and 13 minor uncharacterized isoforms. According to ACMG/AMP-based standards, 15 variants could be classified as pathogenic or likely pathogenic variants: c.404G > A, c.405-6T > A, c.571 + 4A > G, c.571 + 5G > A, c.572-1G > T, c.705G > T, c.706-2A > C, c.706-2A > G, c.837 + 2T > C, c.905-3C > G, c.905-2A > C, c.905-2_905-1del, c.965 + 5G > A, c.1026 + 5_1026 + 7del, and c.1026 + 5G > T.
Collapse
Affiliation(s)
- Lara Sanoguera-Miralles
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas (CSIC-UVa), 47003 Valladolid, Spain; (L.S.-M.); (A.V.-P.); (E.B.-M.); (B.D.-G.); (E.F.-B.)
| | - Alberto Valenzuela-Palomo
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas (CSIC-UVa), 47003 Valladolid, Spain; (L.S.-M.); (A.V.-P.); (E.B.-M.); (B.D.-G.); (E.F.-B.)
| | - Elena Bueno-Martínez
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas (CSIC-UVa), 47003 Valladolid, Spain; (L.S.-M.); (A.V.-P.); (E.B.-M.); (B.D.-G.); (E.F.-B.)
| | - Patricia Llovet
- Molecular Oncology Laboratory CIBERONC, Hospital Clinico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain; (P.L.); (P.P.-S.); (M.C.)
| | - Beatriz Díez-Gómez
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas (CSIC-UVa), 47003 Valladolid, Spain; (L.S.-M.); (A.V.-P.); (E.B.-M.); (B.D.-G.); (E.F.-B.)
| | - María José Caloca
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas (CSIC-UVa), 47003 Valladolid, Spain;
| | - Pedro Pérez-Segura
- Molecular Oncology Laboratory CIBERONC, Hospital Clinico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain; (P.L.); (P.P.-S.); (M.C.)
| | - Eugenia Fraile-Bethencourt
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas (CSIC-UVa), 47003 Valladolid, Spain; (L.S.-M.); (A.V.-P.); (E.B.-M.); (B.D.-G.); (E.F.-B.)
- Knight Cancer Research Building, 2720 S Moody Ave, Portland, OR 97201, USA
| | - Marta Colmena
- Molecular Oncology Laboratory CIBERONC, Hospital Clinico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain; (P.L.); (P.P.-S.); (M.C.)
| | - Sara Carvalho
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK; (S.C.); (J.A.); (D.F.E.)
| | - Jamie Allen
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK; (S.C.); (J.A.); (D.F.E.)
| | - Douglas F. Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK; (S.C.); (J.A.); (D.F.E.)
| | - Peter Devilee
- Leiden University Medical Center, Department of Human Genetics, 2300RC Leiden, The Netherlands; (P.D.); (M.P.G.V.)
| | - Maaike P. G. Vreeswijk
- Leiden University Medical Center, Department of Human Genetics, 2300RC Leiden, The Netherlands; (P.D.); (M.P.G.V.)
| | - Miguel de la Hoya
- Molecular Oncology Laboratory CIBERONC, Hospital Clinico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain; (P.L.); (P.P.-S.); (M.C.)
| | - Eladio A. Velasco
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas (CSIC-UVa), 47003 Valladolid, Spain; (L.S.-M.); (A.V.-P.); (E.B.-M.); (B.D.-G.); (E.F.-B.)
| |
Collapse
|
113
|
Kumari N, Singh RK, Mishra SK, L R, Mohindra S, Krishnani N. Prevalence and spectrum of pathogenic germline variants in intestinal and pancreatobiliary type of ampullary cancer. Pathol Res Pract 2020; 217:153309. [PMID: 33341547 DOI: 10.1016/j.prp.2020.153309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/20/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Ampullary cancer may occur as a component of hereditary cancer syndromes. Mutations in inherited cancer susceptibility genes play a therapeutic role and its knowledge in ampullary cancer is lacking. METHODS Thirty-seven cases of ampullary carcinoma were subjected to tumor-normal whole exome sequencing with mean coverage of 100X (blood) and 200X (tumor). Data were analyzed and correlated with intestinal and pancreatobiliary differentiation. RESULTS There were 22 intestinal, 13 pancreatobiliary and 2 cases of mixed differentiation. One hundred and forty-three germline variations with at least >1 pathogenic germline variants (PGVs) across 83 genes were found in 36 of 37 patients. Twelve genes (14.5 %) showed >3, 20 genes (24.1 %) showed two and 51 genes (61.4 %) showed one PGVs. Intestinal differentiation showed higher PGVs (117 variants, 73 genes) than pancreatobiliary differentiation (85 variants, 62 genes). PGVs in ERCC5, MEN1, MSH3, CHEK1, TP53, APC, FANCA, ERBB2, BRCA1, BRCA2, RTEL1, HNF1A and PTCH1 were seen in >50 % of cases. Nine genes harbored somatic second hits in 14 cases. PGVs in DNA damage-repair, homologous recombination repair, TP53 transcriptional regulation, DNA double stranded breaks, cell cycle and nucleotide excision repair genes were seen in all cases of intestinal and pancreatobiliary differentiation, while DNA mismatch repair genes were found in 81.8 % of intestinal and 84.6 % of pancreatobiliary cancers. Functional pathway analysis showed that DNA damage-repair, double stranded break repair, mismatch repair, homologous recombination repair and TP53 transcriptional regulation genes were altered in both while nucleotide-excision repair was significantly mutated in intestinal type and cell-cycle genes in pancreatobiliary type (p < 0.05). CONCLUSION This study reports spectrum of PGVs in intestinal and pancreatobiliary differentiation of ampullary carcinoma at higher frequency through whole exome sequencing. PGVs were most frequently found in DNA repair genes. Detecting PGVs through tumor-normal sequencing may identify therapeutically actionable and double-hit mutations that can guide towards appropriate management.
Collapse
Affiliation(s)
- Niraj Kumari
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India.
| | - Rajneesh K Singh
- Department of Surgical Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India.
| | - Shravan K Mishra
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India.
| | - Raghvendra L
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India.
| | - Samir Mohindra
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India.
| | - Narendra Krishnani
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India.
| |
Collapse
|
114
|
BRDT promotes ovarian cancer cell growth. Cell Death Dis 2020; 11:1021. [PMID: 33257688 PMCID: PMC7705741 DOI: 10.1038/s41419-020-03225-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 12/18/2022]
Abstract
Bromodomain testis-specific factor (BRDT) is a member of the bromodomain and extra-terminal (BET) family proteins. Its expression and potential functions in ovarian cancer were examined. We show that BRDT is overexpressed in human ovarian cancer tissues and in established (CaOV3)/primary ovarian cancer cells. However, its expression is low in ovarian epithelial tissues and cells. Significantly, shRNA-induced silencing or CRISPR/Cas9-mediated knockout of BRDT inhibited ovarian cancer cell growth, viability, proliferation and migration, and induced significant apoptosis activation. Conversely, exogenous overexpression of BRDT, by a lentiviral construct, augmented CaOV3 cell proliferation and migration. In CaOV3 cells expression of two key BRDT target genes, polo-like kinase 1 (PLK1) and aurora kinase C (AURKC), was downregulated by BRDT shRNA or knockout, but upregulated with BRDT overexpression. In vivo, xenograft tumors-derived from BRDT-knockout CaOV3 cells grew significantly slower than control tumors in severe combined immunodeficient (SCID) mice. Furthermore, intratumoral injection of BRDT shRNA lentivirus potently inhibited the growth of primary ovarian cancer xenografts in SCID mice. Downregulation of PLK1 and AURKC was detected in BRDT-knockout and BRDT-silenced tumor tissues. Collectively, BRDT overexpression promotes ovarian cancer cell progression. Targeting BRDT could be a novel strategy to treat ovarian cancer.
Collapse
|
115
|
Nguyen L, W M Martens J, Van Hoeck A, Cuppen E. Pan-cancer landscape of homologous recombination deficiency. Nat Commun 2020; 11:5584. [PMID: 33149131 PMCID: PMC7643118 DOI: 10.1038/s41467-020-19406-4] [Citation(s) in RCA: 270] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/07/2020] [Indexed: 12/27/2022] Open
Abstract
Homologous recombination deficiency (HRD) results in impaired double strand break repair and is a frequent driver of tumorigenesis. Here, we develop a genome-wide mutational scar-based pan-cancer Classifier of HOmologous Recombination Deficiency (CHORD) that can discriminate BRCA1- and BRCA2-subtypes. Analysis of a metastatic (n = 3,504) and primary (n = 1,854) pan-cancer cohort reveals that HRD is most frequent in ovarian and breast cancer, followed by pancreatic and prostate cancer. We identify biallelic inactivation of BRCA1, BRCA2, RAD51C or PALB2 as the most common genetic cause of HRD, with RAD51C and PALB2 inactivation resulting in BRCA2-type HRD. We find that while the specific genetic cause of HRD is cancer type specific, biallelic inactivation is predominantly associated with loss-of-heterozygosity (LOH), with increased contribution of deep deletions in prostate cancer. Our results demonstrate the value of pan-cancer genomics-based HRD testing and its potential diagnostic value for patient stratification towards treatment with e.g. poly ADP-ribose polymerase inhibitors (PARPi). Cancers deficient in homologous recombination can benefit from treatment with poly ADP-ribose polymerase (PARP) inhibitors. Here, the authors generated a classifier that can predict homologous recombination deficiency from genomic data and suggest several cancer types that may benefit from PARP inhibitor treatment.
Collapse
Affiliation(s)
- Luan Nguyen
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC Cancer institute, Erasmus University Medical Center, Rotterdam, The Netherlands.,Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
| | - Arne Van Hoeck
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Edwin Cuppen
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht, The Netherlands. .,Hartwig Medical Foundation, Amsterdam, The Netherlands.
| |
Collapse
|
116
|
Lang GT, Shi JX, Huang L, Cao AY, Zhang CH, Song CG, Zhuang ZG, Hu X, Huang W, Shao ZM. Multiple cancer susceptible genes sequencing in BRCA-negative breast cancer with high hereditary risk. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1417. [PMID: 33313162 PMCID: PMC7723566 DOI: 10.21037/atm-20-2999] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Hereditary factors contributed to breast cancer susceptibility. Low BRCA mutation prevalence was demonstrated in previous BRCA mutation screening in Chinese breast cancer patients. Multiple-gene sequencing may assist in discovering detrimental germline mutation in BRCA negative breast cancers. Methods A total of 384 Chinese subjects with any two of high-risk factors were recruited and screened by next-generation sequencing (NGS) for 30 cancer susceptible genes. Variants with a truncating, initiation codon or splice donor/acceptor effect, or with pathogenicity demonstrated in published literature were classified into pathogenic/likely-pathogenic mutations. Results In total, we acquired 39 (10.2%) patients with pathogenic/likely-pathogenic germline mutations, including one carrying two distinct mutations. Major mutant non-BRCA genes were MUTYH (n=11, 2.9%), PTCH1 (n=7, 1.8%), RET (n=6, 1.6%) and PALB2 (n=5, 1.3%). Other mutant genes included TP53 (n=3, 0.8%), RAD51D (n=2, 0.5%), CHEK2 (n=1, 0.3%), BRIP1 (n=1, 0.3%), CDH1 (n=1, 0.3%), MRE11 (n=1, 0.3%), RAD50 (n=1, 0.3%) and PALLD (n=1, 0.3%). A splicing germline mutation, MUTYH c.934-2A>G, was a hotspot (9/384, 2.3%) in Chinese breast cancer. Conclusions Among BRCA-negative breast cancer patients with high hereditary risk in China, 10.2% carried mutations in cancer associated susceptibility genes. MUTYH and PTCH1 had relatively high mutation rates (2.9% and 1.8%). Multigene testing contributes to understand genetic background of BRCA-negative breast cancer patients with high hereditary risk.
Collapse
Affiliation(s)
- Guan-Tian Lang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin-Xiu Shi
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Industrial Technology Institute (SITI), Shanghai, China
| | - Liang Huang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - A-Yong Cao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen-Hui Zhang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Industrial Technology Institute (SITI), Shanghai, China
| | - Chuan-Gui Song
- Department of Breast Surgery, Affiliated Union Hospital, Fujian Medical University, Fuzhou, China
| | - Zhi-Gang Zhuang
- Department of Breast Surgery, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xin Hu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Huang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Industrial Technology Institute (SITI), Shanghai, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
117
|
Purrington KS, Raychaudhuri S, Simon MS, Clark J, Ratliff V, Dyson G, Craig DB, Boerner JL, Beebe-Dimmer JL, Schwartz AG. Heritable Susceptibility to Breast Cancer among African-American Women in the Detroit Research on Cancer Survivors Study. Cancer Epidemiol Biomarkers Prev 2020; 29:2369-2375. [PMID: 32868316 PMCID: PMC7642006 DOI: 10.1158/1055-9965.epi-20-0564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/16/2020] [Accepted: 08/26/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND African-American women have high rates of breast cancer associated with hereditary features. However, no studies have reported the prevalence of inherited variation across all genes known to be breast cancer risk factors among African-American patients with breast cancer not selected for high-risk characteristics. METHODS We evaluated 182 African-American women diagnosed with invasive breast cancer in metropolitan Detroit via targeted capture and multiplex sequencing of 13 well-established breast cancer risk genes and five suggested breast cancer risk genes. RESULTS We identified 24 pathogenic variants in 23 women [12.6%; 95% confidence interval (CI), 8.2%-18.4%] and five genes (BRCA2, BRCA1, ATM, RAD50, CDH1). BRCA1 and BRCA2 accounted for 58.3% of all pathogenic variants. An additional six pathogenic variants were found in suggested breast cancer risk genes (MSH6, MUTYH, NF1, BRIP1). CONCLUSIONS The prevalence of germline pathogenic variants is relatively high among African-American patients with breast cancer unselected for high-risk characteristics across a broad spectrum of genes. IMPACT This study helps to define the genomic landscape of breast cancer susceptibility in African-American women who could benefit from enhanced surveillance and screening.
Collapse
Affiliation(s)
- Kristen S Purrington
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | | | - Michael S Simon
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Julie Clark
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Valerie Ratliff
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
| | - Gregory Dyson
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
- Bioinformatics & Biostatistics Core, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Douglas B Craig
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Bioinformatics & Biostatistics Core, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Julie L Boerner
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Jennifer L Beebe-Dimmer
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Ann G Schwartz
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| |
Collapse
|
118
|
Purrington KS, Schwartz AG, Ruterbusch JJ, Manning MA, Nair M, Wenzlaff AS, Pandolfi SS, Simon MS, Beebe-Dimmer J. Patterns of cancer family history and genetic counseling eligibility among African Americans with breast, prostate, lung, and colorectal cancers: A Detroit Research on Cancer Survivors cohort study. Cancer 2020; 126:4744-4752. [PMID: 32749684 PMCID: PMC8027783 DOI: 10.1002/cncr.33126] [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: 05/12/2020] [Revised: 07/02/2020] [Accepted: 07/08/2020] [Indexed: 11/05/2022]
Abstract
BACKGROUND Family history (FH) remains one of the strongest risk factors for many common cancers and is used to determine cancer genetic counseling (CGC) eligibility, but the understanding of familial cancer patterns in African Americans is limited. METHODS This study evaluated cancer FH among African Americans with invasive breast cancer, prostate cancer, lung cancer, or colorectal cancer (CRC) in the Detroit Research on Cancer Survivors (ROCS) cohort. Associations between participant cancer type, site-specific FH, and meeting national guidelines for CGC were evaluated via logistic regression. Cancer FH patterns were evaluating via hierarchical clustering. RESULTS Among 1500 ROCS participants, 71% reported at least 1 first-degree relative or grandparent with cancer. FHs of breast cancer, CRC, lung cancer, and prostate cancer were most common among participants with the same diagnosis (odds ratio [OR] for breast cancer, 1.14; P < .001; OR for CRC, 1.08; P = .003; OR for lung cancer, 1.09; P = .008; OR for prostate cancer, 1.14; P < .001). Nearly half of the participants (47%) met national CGC guidelines, and 24.4% of these participants met CGC criteria on the basis of their cancer FH alone. FH was particularly important in determining CGC eligibility for participants with prostate cancer versus breast cancer (OR for FH vs personal history alone, 2.91; 95% confidence interval, 1.94-4.35; P < .001). In clustering analyses, breast and prostate cancer FH-defined clusters were common across all participants. Clustering of CRC and breast cancer FHs was also observed. CONCLUSIONS ROCS participants reported high rates of cancer FH. The high rate of eligibility for CGC among ROCS participants supports the need for interventions to increase referrals and uptake of CGC among African Americans.
Collapse
Affiliation(s)
- Kristen S. Purrington
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Ann G. Schwartz
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Julie J. Ruterbusch
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Mark A. Manning
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Mrudula Nair
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Angela S. Wenzlaff
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Stephanie S. Pandolfi
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Michael S. Simon
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Jennifer Beebe-Dimmer
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| |
Collapse
|
119
|
Tuazon AMDA, Lott P, Bohórquez M, Benavides J, Ramirez C, Criollo A, Estrada-Florez A, Mateus G, Velez A, Carmona J, Olaya J, Garcia E, Polanco-Echeverry G, Stultz J, Alvarez C, Tapia T, Ashton-Prolla P, Vega A, Lazaro C, Tornero E, Martinez-Bouzas C, Infante M, De La Hoya M, Diez O, Browning BL, Rannala B, Teixeira MR, Carvallo P, Echeverry M, Carvajal-Carmona LG. Haplotype analysis of the internationally distributed BRCA1 c.3331_3334delCAAG founder mutation reveals a common ancestral origin in Iberia. Breast Cancer Res 2020; 22:108. [PMID: 33087180 PMCID: PMC7579869 DOI: 10.1186/s13058-020-01341-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/16/2020] [Indexed: 12/02/2022] Open
Abstract
Background The BRCA1 c.3331_3334delCAAG founder mutation has been reported in hereditary breast and ovarian cancer families from multiple Hispanic groups. We aimed to evaluate BRCA1 c.3331_3334delCAAG haplotype diversity in cases of European, African, and Latin American ancestry. Methods BC mutation carrier cases from Colombia (n = 32), Spain (n = 13), Portugal (n = 2), Chile (n = 10), Africa (n = 1), and Brazil (n = 2) were genotyped with the genome-wide single nucleotide polymorphism (SNP) arrays to evaluate haplotype diversity around BRCA1 c.3331_3334delCAAG. Additional Portuguese (n = 13) and Brazilian (n = 18) BC mutation carriers were genotyped for 15 informative SNPs surrounding BRCA1. Data were phased using SHAPEIT2, and identical by descent regions were determined using BEAGLE and GERMLINE. DMLE+ was used to date the mutation in Colombia and Iberia. Results The haplotype reconstruction revealed a shared 264.4-kb region among carriers from all six countries. The estimated mutation age was ~ 100 generations in Iberia and that it was introduced to South America early during the European colonization period. Conclusions Our results suggest that this mutation originated in Iberia and later introduced to Colombia and South America at the time of Spanish colonization during the early 1500s. We also found that the Colombian mutation carriers had higher European ancestry, at the BRCA1 gene harboring chromosome 17, than controls, which further supported the European origin of the mutation. Understanding founder mutations in diverse populations has implications in implementing cost-effective, ancestry-informed screening.
Collapse
Affiliation(s)
| | - Paul Lott
- Genome Center, University of California Davis, Davis, CA, USA
| | | | | | | | | | | | | | - Alejandro Velez
- Hospital Pablo Tobon Uribe, Medellín, Colombia.,Dinamica IPS, Medellín, Colombia
| | | | - Justo Olaya
- Hospital Universitario Hernando Moncaleano Perdomo, Neiva, Colombia
| | - Elisha Garcia
- Genome Center, University of California Davis, Davis, CA, USA
| | | | - Jacob Stultz
- Genome Center, University of California Davis, Davis, CA, USA
| | | | - Teresa Tapia
- Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricia Ashton-Prolla
- Department of Genetics, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Post-graduate Course in Genetics and Molecular Biology, UFRGS, Porto Alegre, Brazil.,Medical Genetics Service, Hospital de Clinicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | | | - Ana Vega
- Fundación Pública Galega de Medicina Xenómica, Grupo de Medicina Xenómica-USC, CIBERER, IDIS, Santiago de Compostela, Spain
| | - Conxi Lazaro
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Eva Tornero
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | | | - Mar Infante
- Cancer Genetics Group, Institute of Genetics and Molecular Biology (UVa-CSIC), Valladolid, Spain
| | - Miguel De La Hoya
- Laboratorio de Oncología Molecular, Hospital Clínico San Carlos. IdISSC (Instituto de Investigación Sanitaria San Carlos), Madrid, Spain
| | - Orland Diez
- Grupo de Cáncer Hereditario, Instituto Oncológico Vall d'Hebron (VHIO), Madrid, Spain
| | - Brian L Browning
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | | | - Bruce Rannala
- Department of Evolution and Ecology, University of California Davis, Davis, CA, USA
| | - Manuel R Teixeira
- Portuguese Oncology Institute of Porto (IPO Porto) and Biomedical Sciences Institute (ICBAS), University of Porto, Porto, Portugal
| | - Pilar Carvallo
- Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Luis G Carvajal-Carmona
- Genome Center, University of California Davis, Davis, CA, USA. .,Division de Investigaciones, Fundacion de Genética y Genómica, Ibague, Colombia. .,University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA. .,Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA.
| |
Collapse
|
120
|
Cavaillé M, Uhrhammer N, Privat M, Ponelle-Chachuat F, Gay-Bellile M, Lepage M, Viala S, Bidet Y, Bignon YJ. Feedback of extended panel sequencing in 1530 patients referred for suspicion of hereditary predisposition to adult cancers. Clin Genet 2020; 99:166-175. [PMID: 33047316 PMCID: PMC7821123 DOI: 10.1111/cge.13864] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 12/12/2022]
Abstract
High‐throughput sequencing analysis represented both a medical diagnosis and technological revolution. Gene panel analysis is now routinely performed in the exploration of hereditary predisposition to cancer, which is becoming increasingly heterogeneous, both clinically and molecularly. We present 1530 patients with suspicion of hereditary predisposition to cancer, for which two types of analyses were performed: a) oriented according to the clinical presentation (n = 417), or b) extended to genes involved in hereditary predisposition to adult cancer (n = 1113). Extended panel analysis had a higher detection rate compared to oriented analysis in hereditary predisposition to breast / ovarian cancer (P < .001) and in digestive cancers (P < .094) (respectively 15% vs 5% and 19.3%, vs 12.5%). This higher detection is explained by the inclusion of moderate penetrance genes, as well as the identification of incident mutations and double mutations. Our study underscores the utility of proposing extended gene panel analysis to patients with suspicion of hereditary predisposition to adult cancer.
Collapse
Affiliation(s)
- Mathias Cavaillé
- Département d'Oncogénétique, Centre Jean Perrin, Clermont-Ferrand, France.,Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies, Clermont Ferrand, France
| | - Nancy Uhrhammer
- Département d'Oncogénétique, Centre Jean Perrin, Clermont-Ferrand, France.,Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies, Clermont Ferrand, France
| | - Maud Privat
- Département d'Oncogénétique, Centre Jean Perrin, Clermont-Ferrand, France.,Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies, Clermont Ferrand, France
| | - Flora Ponelle-Chachuat
- Département d'Oncogénétique, Centre Jean Perrin, Clermont-Ferrand, France.,Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies, Clermont Ferrand, France
| | - Mathilde Gay-Bellile
- Département d'Oncogénétique, Centre Jean Perrin, Clermont-Ferrand, France.,Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies, Clermont Ferrand, France
| | - Mathis Lepage
- Département d'Oncogénétique, Centre Jean Perrin, Clermont-Ferrand, France
| | - Sandrine Viala
- Département d'Oncogénétique, Centre Jean Perrin, Clermont-Ferrand, France.,Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies, Clermont Ferrand, France
| | - Yannick Bidet
- Département d'Oncogénétique, Centre Jean Perrin, Clermont-Ferrand, France.,Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies, Clermont Ferrand, France
| | - Yves-Jean Bignon
- Département d'Oncogénétique, Centre Jean Perrin, Clermont-Ferrand, France.,Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies, Clermont Ferrand, France
| |
Collapse
|
121
|
Tobalina L, Armenia J, Irving E, O'Connor MJ, Forment JV. A meta-analysis of reversion mutations in BRCA genes identifies signatures of DNA end-joining repair mechanisms driving therapy resistance. Ann Oncol 2020; 32:103-112. [PMID: 33091561 DOI: 10.1016/j.annonc.2020.10.470] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/27/2020] [Accepted: 10/04/2020] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Germline mutations in the BRCA1 or BRCA2 (BRCA) genes predispose to hereditary breast and ovarian cancer and, mostly in the case of BRCA2, are also prevalent in cases of pancreatic and prostate malignancies. Tumours from these patients tend to lose both copies of the wild-type BRCA gene, which makes them exquisitely sensitive to platinum drugs and poly(ADP-ribose) polymerase inhibitors (PARPi), treatments of choice in these disease settings. Reversion secondary mutations with the capacity of restoring BRCA protein expression have been documented in the literature as bona fide mechanisms of resistance to these treatments. PATIENTS AND METHODS We analysed published sequencing data of BRCA genes (from tumour or circulating tumour DNA) in 327 patients with tumours harbouring mutations in BRCA1 or BRCA2 (234 patients with ovarian cancer, 27 with breast cancer, 13 with pancreatic cancer, 11 with prostate cancer and 42 with a cancer of unknown origin) that progressed on platinum or PARPi treatment. RESULTS We describe 269 cases of reversion mutations in 86 patients in this cohort (26.0%). Detailed analyses of the reversion events highlight that most amino acid sequences encoded by exon 11 in BRCA1 and BRCA2 are dispensable to generate resistance to platinum or PARPi, whereas other regions are more refractory to sizeable amino acid losses. They also underline the key role of mutagenic end-joining DNA repair pathways in generating reversions, especially in those affecting BRCA2, as indicated by the significant accumulation of DNA sequence microhomologies surrounding deletions leading to reversion events. CONCLUSIONS Our analyses suggest that pharmacological inhibition of DNA end-joining repair pathways could improve durability of drug treatments by preventing the acquisition of reversion mutations in BRCA genes. They also highlight potential new therapeutic opportunities when reversions result in expression of hypomorphic versions of BRCA proteins, especially with agents targeting the response to DNA replication stress.
Collapse
Affiliation(s)
- L Tobalina
- Bioinformatics and Data Science, Oncology R&D, AstraZeneca, Cambridge, UK
| | - J Armenia
- Bioinformatics and Data Science, Oncology R&D, AstraZeneca, Cambridge, UK
| | - E Irving
- DDR Biology, Bioscience, Oncology R&D, AstraZeneca, Cambridge, UK
| | - M J O'Connor
- DDR Biology, Bioscience, Oncology R&D, AstraZeneca, Cambridge, UK
| | - J V Forment
- DDR Biology, Bioscience, Oncology R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
122
|
Inagaki-Kawata Y, Yoshida K, Kawaguchi-Sakita N, Kawashima M, Nishimura T, Senda N, Shiozawa Y, Takeuchi Y, Inoue Y, Sato-Otsubo A, Fujii Y, Nannya Y, Suzuki E, Takada M, Tanaka H, Shiraishi Y, Chiba K, Kataoka Y, Torii M, Yoshibayashi H, Yamagami K, Okamura R, Moriguchi Y, Kato H, Tsuyuki S, Yamauchi A, Suwa H, Inamoto T, Miyano S, Ogawa S, Toi M. Genetic and clinical landscape of breast cancers with germline BRCA1/2 variants. Commun Biol 2020; 3:578. [PMID: 33067557 PMCID: PMC7567851 DOI: 10.1038/s42003-020-01301-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 09/15/2020] [Indexed: 12/24/2022] Open
Abstract
The genetic and clinical characteristics of breast tumors with germline variants, including their association with biallelic inactivation through loss-of-heterozygosity (LOH) and second somatic mutations, remain elusive. We analyzed germline variants of 11 breast cancer susceptibility genes for 1,995 Japanese breast cancer patients, and identified 101 (5.1%) pathogenic variants, including 62 BRCA2 and 15 BRCA1 mutations. Genetic analysis of 64 BRCA1/2-mutated tumors including TCGA dataset tumors, revealed an association of biallelic inactivation with more extensive deletions, copy neutral LOH, gain with LOH and younger onset. Strikingly, TP53 and RB1 mutations were frequently observed in BRCA1- (94%) and BRCA2- (9.7%) mutated tumors with biallelic inactivation. Inactivation of TP53 and RB1 together with BRCA1 and BRCA2, respectively, involved LOH of chromosomes 17 and 13. Notably, BRCA1/2 tumors without biallelic inactivation were indistinguishable from those without germline variants. Our study highlights the heterogeneity and unique clonal selection pattern in breast cancers with germline variants. Yukiko Inagaki-Kawata et al. report an analysis of germline variants in breast cancer susceptibility genes in 1,995 Japanese breast cancer patients. They find that 5.1% of the patients carry germline variants in cancer-linked genes and investigate the characteristics of patients with germline mutations in BRCA1/2.
Collapse
Affiliation(s)
- Yukiko Inagaki-Kawata
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan.,Department of Breast Surgery, Kyoto University, Kyoto, Japan
| | - Kenichi Yoshida
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | | | | | - Tomomi Nishimura
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan.,Department of Breast Surgery, Kyoto University, Kyoto, Japan
| | - Noriko Senda
- Department of Breast Surgery, Kyoto University, Kyoto, Japan
| | - Yusuke Shiozawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Yasuhide Takeuchi
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan.,Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan.,Department of Diagnostic Pathology, Kyoto University, Kyoto, Japan
| | - Yoshikage Inoue
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Aiko Sato-Otsubo
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Yoichi Fujii
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Yasuhito Nannya
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Eiji Suzuki
- Department of Breast Surgery, Kyoto University, Kyoto, Japan
| | - Masahiro Takada
- Department of Breast Surgery, Kyoto University, Kyoto, Japan
| | - Hiroko Tanaka
- Laboratory of Sequence Analysis, Human Genome Centre, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuichi Shiraishi
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kenichi Chiba
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuki Kataoka
- Hospital Care Research Unit/Department of Respiratory Medicine, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Japan
| | - Masae Torii
- Department of Breast Surgery, Japanese Red Cross Wakayama Medical Center, Wakayama, Japan
| | - Hiroshi Yoshibayashi
- Department of Breast Surgery, Japanese Red Cross Wakayama Medical Center, Wakayama, Japan
| | | | - Ryuji Okamura
- Department of Breast Surgery, Yamatotakada Municipal Hospital, Yamatotakada, Japan
| | | | - Hironori Kato
- Department of Breast Surgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Shigeru Tsuyuki
- Department of Breast Surgery, Osaka Red Cross Hospital, Osaka, Japan
| | - Akira Yamauchi
- Department of Breast Surgery, Kitano Hospital, Osaka, Japan
| | - Hirofumi Suwa
- Department of Breast Surgery, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Japan
| | | | - Satoru Miyano
- Laboratory of Sequence Analysis, Human Genome Centre, Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan. .,Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan. .,Department of Medicine, Centre for Haematology and Regenerative Medicine, Karolinska Institute, Stockholm, Sweden.
| | - Masakazu Toi
- Department of Breast Surgery, Kyoto University, Kyoto, Japan.
| |
Collapse
|
123
|
Grasel RS, Felicio PS, de Paula AE, Campacci N, Garcia FADO, de Andrade ES, Evangelista AF, Fernandes GC, Sabato CDS, De Marchi P, Souza CDP, de Paula CAA, Torrezan GT, Galvão HDCR, Carraro DM, Palmero EI. Using Co-segregation and Loss of Heterozygosity Analysis to Define the Pathogenicity of Unclassified Variants in Hereditary Breast Cancer Patients. Front Oncol 2020; 10:571330. [PMID: 33134171 PMCID: PMC7566163 DOI: 10.3389/fonc.2020.571330] [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: 06/10/2020] [Accepted: 08/28/2020] [Indexed: 12/18/2022] Open
Abstract
The use of gene panels introduces a new dilemma in the genetics field due to the high frequency of variants of uncertain significance (VUS). The objective of this study was to provide evidence that may help in the classification of these germline variants in terms of their clinical impact and association with the disease in question. A total of 52 unrelated women at-risk for HBOC and negative for BRCA1/BRCA2 pathogenic variants were evaluated through a gene panel comprising 14 breast and/or ovarian cancer susceptibility genes. Of the 453 germline variants identified, 15 variants (classes 3, 4, and 5) in the ATM, BRIP1, CHEK2, MRE11A, MUTHY, PALB2, RAD50, and RAD51C genes were evaluated via databases, co-segregation studies and loss of heterozygosity in the tumor. The co-segregation analysis allowed the establishment of an association with the presence of variants and the risk of cancer for variant c.316C>T in the BRIP1 gene. Four variants of uncertain significance showed loss of heterozygosity in the tumor (ATM c.4709T>C; CHEK2 c.1036C>T; PALB2 c.1001A>G, and RAD50 c.281T>C), which is an indication of pathogenicity. Thus, the present study provides novel evidence that favors the association of variants in moderate-risk genes with the development of hereditary breast cancer.
Collapse
Affiliation(s)
| | - Paula Silva Felicio
- Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo, Brazil
| | | | - Natalia Campacci
- Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo, Brazil
| | | | | | | | | | | | - Pedro De Marchi
- Department of Medical Oncology, Barretos Cancer Hospital, São Paulo, Brazil.,Oncoclinicas, Rio de Janeiro, Brazil
| | - Cristiano de Pádua Souza
- Department of Medical Oncology, Barretos Cancer Hospital, São Paulo, Brazil.,Department of Oncogenetics, Barretos Cancer Hospital, São Paulo, Brazil
| | | | | | | | - Dirce Maria Carraro
- Genomic Diagnostic Center, AC Camargo Cancer Center, São Paulo, Brazil.,Genomics and Molecular Biology Group, CIPE - A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Edenir Inêz Palmero
- Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo, Brazil.,Center of Molecular Diagnosis, Barretos Cancer Hospital, São Paulo, Brazil.,Pele Little Prince Research Institute, Curitiba, Brazil.,Faculdades Pequeno Príncipe, Curitiba, Brazil
| |
Collapse
|
124
|
Bandeira G, Rocha K, Lazar M, Ezquina S, Yamamoto G, Varela M, Takahashi V, Aguena M, Gollop T, Zatz M, Passos-Bueno MR, Krepischi A, Okamoto OK. Germline variants of Brazilian women with breast cancer and detection of a novel pathogenic ATM deletion in early-onset breast cancer. Breast Cancer 2020; 28:346-354. [PMID: 32986223 DOI: 10.1007/s12282-020-01165-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/16/2020] [Indexed: 04/07/2023]
Abstract
BACKGROUND It is estimated that 5-10% of breast cancer cases are hereditary. The identification of pathogenic germline variants allows individualized preventive health care, improvement of clinical management and genetic counseling. Studies in ethnically admixed Latin American populations have identified regions with increased frequency of deleterious variants in breast cancer predisposing genes. In this context, the Brazilian population exhibits great genetic heterogeneity, and is not well represented in international databases, which makes it difficult to interpret the clinical relevance of germline variants. METHODS We evaluated the frequency of pathogenic/likely pathogenic (P/LP) germline variants in up to 37 breast cancer predisposing genes, in a cohort of 105 breast and/or ovarian cancer Brazilian women referred to two research centers between 2014 and 2019. RESULTS A total of 22 patients (21%) were found to carry P/LP variants, and 16 VUS were detected in 15 patients (14.3%). Additionally, a novel pathogenic ATM intragenic deletion was identified in an early-onset breast cancer. We also detected a BRCA1 pathogenic variant (c.5074+2T>C) in higher frequency (10×) than in other studies with similar cohorts. CONCLUSIONS Our findings contribute to the characterization of the genetic background of breast cancer predisposition in the Brazilian population as a useful resource to discriminate between deleterious variants and VUS, thus enabling improvement in the preventive health care and clinical management of carriers.
Collapse
Affiliation(s)
- Gabriel Bandeira
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco, Universidade de São Paulo, Cidade Universitária, Rua do Matão 277, São Paulo, SP, CEP: 05508-090, Brazil
| | - Katia Rocha
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco, Universidade de São Paulo, Cidade Universitária, Rua do Matão 277, São Paulo, SP, CEP: 05508-090, Brazil
| | - Monize Lazar
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco, Universidade de São Paulo, Cidade Universitária, Rua do Matão 277, São Paulo, SP, CEP: 05508-090, Brazil
| | - Suzana Ezquina
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco, Universidade de São Paulo, Cidade Universitária, Rua do Matão 277, São Paulo, SP, CEP: 05508-090, Brazil
| | - Guilherme Yamamoto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco, Universidade de São Paulo, Cidade Universitária, Rua do Matão 277, São Paulo, SP, CEP: 05508-090, Brazil.,Genetics Unit, Faculty of Medicine, Children's Institute, Clinics Hospital, University of Sao Paulo, São Paulo, Brazil
| | - Monica Varela
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco, Universidade de São Paulo, Cidade Universitária, Rua do Matão 277, São Paulo, SP, CEP: 05508-090, Brazil
| | - Vanessa Takahashi
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco, Universidade de São Paulo, Cidade Universitária, Rua do Matão 277, São Paulo, SP, CEP: 05508-090, Brazil
| | - Meire Aguena
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco, Universidade de São Paulo, Cidade Universitária, Rua do Matão 277, São Paulo, SP, CEP: 05508-090, Brazil
| | - Thomaz Gollop
- Department of Gynecology and Obstetrics, Faculty of Medicine of Jundiai, São Paulo, Brazil
| | - Mayana Zatz
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco, Universidade de São Paulo, Cidade Universitária, Rua do Matão 277, São Paulo, SP, CEP: 05508-090, Brazil
| | - Maria Rita Passos-Bueno
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco, Universidade de São Paulo, Cidade Universitária, Rua do Matão 277, São Paulo, SP, CEP: 05508-090, Brazil
| | - Ana Krepischi
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco, Universidade de São Paulo, Cidade Universitária, Rua do Matão 277, São Paulo, SP, CEP: 05508-090, Brazil
| | - Oswaldo Keith Okamoto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Centro de Pesquisa Sobre o Genoma Humano e Células-Tronco, Universidade de São Paulo, Cidade Universitária, Rua do Matão 277, São Paulo, SP, CEP: 05508-090, Brazil.
| |
Collapse
|
125
|
Zhang L, Liu Q, Mu Q, Zhou D, Li H, Zhang B, Yin C. MiR-429 suppresses proliferation and invasion of breast cancer via inhibiting the Wnt/β-catenin signaling pathway. Thorac Cancer 2020; 11:3126-3138. [PMID: 32961031 PMCID: PMC7606009 DOI: 10.1111/1759-7714.13620] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 12/24/2022] Open
Abstract
Background microRNAs (miRNAs) have been verified as molecular targets for regulating tumor proliferation, invasion, and metastasis in tumor progression. However, the relationship between miRNAs and cellular energy metabolism in breast cancer still needs to be clarified. This study aimed to investigate the role of miR‐429 in breast cancer progression. Methods Bioinformatic analyses were employed to detect the relationship between miR‐429 and cancer‐related signaling pathways. We used a Kaplan‐Meier curve to analyze survival rate in patients with high or low expression of miR‐429. We used real‐time quantitative PCR (RT‐qPCR) to detect the expression of miR‐429 in different cell lines. Sh‐con, over‐miR‐429, miR‐429 inhibitor, and sh‐inhibitor control were transfected. Colony formation and EDU assay were used to detect the proliferation of transfected cells. Wound healing and transwell assays were performed to detect the mobility and invasion ability of transfected cells. Western blot assay was used to detect relative protein expression in transfected cells and different tissues. Bioinformatic analyses were conducted to detect the target proteins expression in different breast cancer databases. Dual luciferase reporter assay was used to confirm the binding site between miR‐429 and fibronectin 1 (FN1). Results The results of our study indicate that MiR‐429 and its target genes are associated with cancer‐related signaling pathways and that higher miR‐429 expression corresponds with a better prognosis. When miR‐429 was overexpressed, the proliferation, invasion of MDA‐MB‐231 were inhibited. MiR‐429 was able to suppress the Wnt/β‐catenin signaling pathway, and FN1 overexpression could rescue the influence of over‐miR‐429. Conclusions The results of our study suggest that miR‐429 suppresses the proliferation and invasion of breast cancer via inhibiting the Wnt/β‐catenin signaling pathway.
Collapse
Affiliation(s)
- Liping Zhang
- Department of Pathology, Basic Medical College, Weifang Medical University, Weifang, China
| | - Qinghua Liu
- Department of Human Anatomy, Basic Medical College, Weifang Medical University, Weifang, China
| | - Qingjie Mu
- Department of Oncology, Clinical Medical College, Weifang Medical University, Weifang, China
| | - Dandan Zhou
- Department of Pathology, Basic Medical College, Weifang Medical University, Weifang, China
| | - Hongli Li
- Medicine Research Center, Weifang Medical University, Weifang, China
| | - Baogang Zhang
- Department of Pathology, Basic Medical College, Weifang Medical University, Weifang, China
| | - Chonggao Yin
- College of Nursing, Weifang Medical University, Weifang, China
| |
Collapse
|
126
|
t'Kint de Roodenbeke MD, Pondé N, Buisseret L, Piccart M. Management of early breast cancer in patients bearing germline BRCA mutations. Semin Oncol 2020; 47:243-248. [PMID: 32912765 DOI: 10.1053/j.seminoncol.2020.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 07/27/2020] [Accepted: 07/31/2020] [Indexed: 11/11/2022]
Abstract
Women diagnosed with breast cancers (BCs) that harbor BRCA1/2 mutations have an increased lifetime risk of a second BC and ovarian cancer. They may benefit from risk-reducing surgical strategies such as mastectomy and salpingo-oophorectomy. In cases of triple negative BC with BRCA mutation, there is some evidence that adding platinum-agents in the neoadjuvant setting improves the pathologic complete response. Lastly, ongoing clinical trials testing the efficacy of PARP inhibitor therapy in tumors with BRCA1/2 mutations will be determinant for future guideline recommendations in selecting best adjuvant treatment options for this specific population. For pre-menopausal patients whose tumors have BRCA mutations and hormone-receptor positive BC, the option of combined bilateral annexectomy and hormonal therapy with aromatase inhibitor can be discussed with high-risk patients. This review summarizes the latest results from clinical trials evaluating treatment and prevention strategies for breast cancers harboring BRCA1/2 mutations and discusses the current management of this patient population.
Collapse
Affiliation(s)
| | - Noam Pondé
- Oncology Department AC Camargo Cancer Center, Sao Paulo, Brazil
| | - Laurence Buisseret
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Martine Piccart
- Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
127
|
Lodovichi S, Cervelli T, Pellicioli A, Galli A. Inhibition of DNA Repair in Cancer Therapy: Toward a Multi-Target Approach. Int J Mol Sci 2020; 21:E6684. [PMID: 32932697 PMCID: PMC7554826 DOI: 10.3390/ijms21186684] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/19/2022] Open
Abstract
Alterations in DNA repair pathways are one of the main drivers of cancer insurgence. Nevertheless, cancer cells are more susceptible to DNA damage than normal cells and they rely on specific functional repair pathways to survive. Thanks to advances in genome sequencing, we now have a better idea of which genes are mutated in specific cancers and this prompted the development of inhibitors targeting DNA repair players involved in pathways essential for cancer cells survival. Currently, the pivotal concept is that combining the inhibition of mechanisms on which cancer cells viability depends is the most promising way to treat tumorigenesis. Numerous inhibitors have been developed and for many of them, efficacy has been demonstrated either alone or in combination with chemo or radiotherapy. In this review, we will analyze the principal pathways involved in cell cycle checkpoint and DNA repair focusing on how their alterations could predispose to cancer, then we will explore the inhibitors developed or in development specifically targeting different proteins involved in each pathway, underscoring the rationale behind their usage and how their combination and/or exploitation as adjuvants to classic therapies could help in patients clinical outcome.
Collapse
Affiliation(s)
- Samuele Lodovichi
- Bioscience Department, University of Milan, Via Celoria 26, 20131 Milan, Italy;
| | - Tiziana Cervelli
- Yeast Genetics and Genomics Group, Laboratory of Functional Genetics and Genomics, Institute of Clinical Physiology CNR, Via Moruzzi 1, 56125 Pisa, Italy;
| | - Achille Pellicioli
- Bioscience Department, University of Milan, Via Celoria 26, 20131 Milan, Italy;
| | - Alvaro Galli
- Yeast Genetics and Genomics Group, Laboratory of Functional Genetics and Genomics, Institute of Clinical Physiology CNR, Via Moruzzi 1, 56125 Pisa, Italy;
| |
Collapse
|
128
|
Aganezov S, Goodwin S, Sherman RM, Sedlazeck FJ, Arun G, Bhatia S, Lee I, Kirsche M, Wappel R, Kramer M, Kostroff K, Spector DL, Timp W, McCombie WR, Schatz MC. Comprehensive analysis of structural variants in breast cancer genomes using single-molecule sequencing. Genome Res 2020; 30:1258-1273. [PMID: 32887686 PMCID: PMC7545150 DOI: 10.1101/gr.260497.119] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 08/07/2020] [Indexed: 12/14/2022]
Abstract
Improved identification of structural variants (SVs) in cancer can lead to more targeted and effective treatment options as well as advance our basic understanding of the disease and its progression. We performed whole-genome sequencing of the SKBR3 breast cancer cell line and patient-derived tumor and normal organoids from two breast cancer patients using Illumina/10x Genomics, Pacific Biosciences (PacBio), and Oxford Nanopore Technologies (ONT) sequencing. We then inferred SVs and large-scale allele-specific copy number variants (CNVs) using an ensemble of methods. Our findings show that long-read sequencing allows for substantially more accurate and sensitive SV detection, with between 90% and 95% of variants supported by each long-read technology also supported by the other. We also report high accuracy for long reads even at relatively low coverage (25×–30×). Furthermore, we integrated SV and CNV data into a unifying karyotype-graph structure to present a more accurate representation of the mutated cancer genomes. We find hundreds of variants within known cancer-related genes detectable only through long-read sequencing. These findings highlight the need for long-read sequencing of cancer genomes for the precise analysis of their genetic instability.
Collapse
Affiliation(s)
- Sergey Aganezov
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21211, USA
| | - Sara Goodwin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Rachel M Sherman
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21211, USA
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Gayatri Arun
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Sonam Bhatia
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Isac Lee
- Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21211, USA
| | - Melanie Kirsche
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21211, USA
| | - Robert Wappel
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Melissa Kramer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | | | - David L Spector
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Winston Timp
- Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21211, USA
| | | | - Michael C Schatz
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21211, USA.,Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA.,Department of Biology, Johns Hopkins University, Baltimore, Maryland 21211, USA
| |
Collapse
|
129
|
Gruber JJ, Chen J, Geller B, Jäger N, Lipchik AM, Wang G, Kurian AW, Ford JM, Snyder MP. Chromatin Remodeling in Response to BRCA2-Crisis. Cell Rep 2020; 28:2182-2193.e6. [PMID: 31433991 DOI: 10.1016/j.celrep.2019.07.057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 06/05/2019] [Accepted: 07/17/2019] [Indexed: 10/26/2022] Open
Abstract
Individuals with a single functional copy of the BRCA2 tumor suppressor have elevated risks for breast, ovarian, and other solid tumor malignancies. The exact mechanisms of carcinogenesis due to BRCA2 haploinsufficiency remain unclear, but one possibility is that at-risk cells are subject to acute periods of decreased BRCA2 availability and function ("BRCA2-crisis"), which may contribute to disease. Here, we establish an in vitro model for BRCA2-crisis that demonstrates chromatin remodeling and activation of an NF-κB survival pathway in response to transient BRCA2 depletion. Mechanistically, we identify BRCA2 chromatin binding, histone acetylation, and associated transcriptional activity as critical determinants of the epigenetic response to BRCA2-crisis. These chromatin alterations are reflected in transcriptional profiles of pre-malignant tissues from BRCA2 carriers and, therefore, may reflect natural steps in human disease. By modeling BRCA2-crisis in vitro, we have derived insights into pre-neoplastic molecular alterations that may enhance the development of preventative therapies.
Collapse
Affiliation(s)
- Joshua J Gruber
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Oncology Division, Stanford University, Stanford, CA 94305, USA
| | - Justin Chen
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Benjamin Geller
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Natalie Jäger
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Andrew M Lipchik
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Guangwen Wang
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Allison W Kurian
- Department of Medicine, Oncology Division, Stanford University, Stanford, CA 94305, USA
| | - James M Ford
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Oncology Division, Stanford University, Stanford, CA 94305, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
130
|
Park K, Kim MK, Lee T, Hong J, Kim H, Ahn S, Lee Y, Kim J, Lee S, Lee JW, Lee W, Chun S, Son BH, Jung KH, Kim Y, Min W, Ahn S. Performance evaluation of an amplicon-based next-generation sequencing panel for BRCA1 and BRCA2 variant detection. J Clin Lab Anal 2020; 34:e23524. [PMID: 32812259 PMCID: PMC7755770 DOI: 10.1002/jcla.23524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/16/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND As next-generation sequencing (NGS) technology matures, various amplicon-based NGS tests for BRCA1/2 genotyping have been introduced. This study was designed to evaluate an NGS test using a newly released amplicon-based panel, AmpliSeq for Illumina BRCA Panel (AmpliSeq panel), for detection of clinically significant BRCA variants, and to compare it to another amplicon-based NGS test confirmed by Sanger sequencing. METHODS We reviewed BRCA test results done by NGS using the TruSeq Custom Amplicon kit from patients suspected of hereditary breast/ovarian cancer syndrome (HBOC) in 2018. Of those, 96 residual samples with 100 clinically significant variants were included in this study using predefined criteria: 100 variants were distributed throughout the BRCA1 and BRCA2 genes. All target variants were confirmed by Sanger sequencing. Duplicate NGS testing of these samples was performed using the AmpliSeq panel, and the concordance of results from the two amplicon-based NGS tests was assessed. RESULTS Ninety-nine of 100 variants were detected in duplicate BRCA1/2 genotyping using the AmpliSeq panel (sensitivity, 99%; specificity, 100%). In the discordant case, one variant (BRCA1 c.3627dupA) was found only in repeat 1, but not in repeat 2. Automated nomenclature of all variants, except for two indel variants, was in consensus with Human Genome Variation Society nomenclature. CONCLUSION Our findings confirm that the analytic performance of the AmpliSeq panel is satisfactory, with high sensitivity and specificity.
Collapse
Affiliation(s)
- Kuenyoul Park
- Department of Laboratory Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Min Kyu Kim
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Samsung Changwon HospitalSungkyunkwan University School of MedicineChangwon‐SiKorea
| | - Taegeun Lee
- Department of Laboratory Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Jinyoung Hong
- Department of Laboratory Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Hyun‐Ki Kim
- Department of Laboratory Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Sunyoung Ahn
- Department of Laboratory MedicineKangwon National University School of MedicineChuncheonKorea
| | - Young‐Jae Lee
- Department of Obstetrics and Gynecology, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Jisun Kim
- Department of Surgery, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Shin‐Wha Lee
- Department of Obstetrics and Gynecology, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Jong Won Lee
- Department of Surgery, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Woochang Lee
- Department of Laboratory Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Sail Chun
- Department of Laboratory Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Byung Ho Son
- Department of Surgery, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Kyung Hae Jung
- Department of Oncology, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Yong‐Man Kim
- Department of Obstetrics and Gynecology, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Won‐Ki Min
- Department of Laboratory Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Sei‐Hyun Ahn
- Department of Surgery, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| |
Collapse
|
131
|
DNA Mismatch Repair Gene Variants in Sporadic Solid Cancers. Int J Mol Sci 2020; 21:ijms21155561. [PMID: 32756484 PMCID: PMC7432688 DOI: 10.3390/ijms21155561] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/18/2022] Open
Abstract
The phenotypic effects of single nucleotide polymorphisms (SNPs) in the development of sporadic solid cancers are still scarce. The aim of this review was to summarise and analyse published data on the associations between SNPs in mismatch repair genes and various cancers. The mismatch repair system plays a unique role in the control of the genetic integrity and it is often inactivated (germline and somatic mutations and hypermethylation) in cancer patients. Here, we focused on germline variants in mismatch repair genes and found the outcomes rather controversial: some SNPs are sometimes ascribed as protective, while other studies reported their pathological effects. Regarding the complexity of cancer as one disease, we attempted to ascertain if particular polymorphisms exert the effect in the same direction in the development and treatment of different malignancies, although it is still not straightforward to conclude whether polymorphisms always play a clear positive role or a negative one. Most recent and robust genome-wide studies suggest that risk of cancer is modulated by variants in mismatch repair genes, for example in colorectal cancer. Our study shows that rs1800734 in MLH1 or rs2303428 in MSH2 may influence the development of different malignancies. The lack of functional studies on many DNA mismatch repair SNPs as well as their interactions are not explored yet. Notably, the concerted action of more variants in one individual may be protective or harmful. Further, complex interactions of DNA mismatch repair variations with both the environment and microenvironment in the cancer pathogenesis will deserve further attention.
Collapse
|
132
|
Alenezi WM, Fierheller CT, Recio N, Tonin PN. Literature Review of BARD1 as a Cancer Predisposing Gene with a Focus on Breast and Ovarian Cancers. Genes (Basel) 2020; 11:E856. [PMID: 32726901 PMCID: PMC7464855 DOI: 10.3390/genes11080856] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/19/2022] Open
Abstract
Soon after the discovery of BRCA1 and BRCA2 over 20 years ago, it became apparent that not all hereditary breast and/or ovarian cancer syndrome families were explained by germline variants in these cancer predisposing genes, suggesting that other such genes have yet to be discovered. BRCA1-associated ring domain (BARD1), a direct interacting partner of BRCA1, was one of the earliest candidates investigated. Sequencing analyses revealed that potentially pathogenic BARD1 variants likely conferred a low-moderate risk to hereditary breast cancer, but this association is inconsistent. Here, we review studies of BARD1 as a cancer predisposing gene and illustrate the challenge of discovering additional cancer risk genes for hereditary breast and/or ovarian cancer. We selected peer reviewed research articles that focused on three themes: (i) sequence analyses of BARD1 to identify potentially pathogenic germline variants in adult hereditary cancer syndromes; (ii) biological assays of BARD1 variants to assess their effect on protein function; and (iii) association studies of BARD1 variants in family-based and case-control study groups to assess cancer risk. In conclusion, BARD1 is likely to be a low-moderate penetrance breast cancer risk gene.
Collapse
Affiliation(s)
- Wejdan M. Alenezi
- Department of Human Genetics, McGill University, Montreal, QC H3A 0G4, Canada; (W.M.A.); (C.T.F.); (N.R.)
- Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Medical Laboratory Technology, Taibah University, Medina 42353, Saudi Arabia
| | - Caitlin T. Fierheller
- Department of Human Genetics, McGill University, Montreal, QC H3A 0G4, Canada; (W.M.A.); (C.T.F.); (N.R.)
- Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Neil Recio
- Department of Human Genetics, McGill University, Montreal, QC H3A 0G4, Canada; (W.M.A.); (C.T.F.); (N.R.)
- Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Patricia N. Tonin
- Department of Human Genetics, McGill University, Montreal, QC H3A 0G4, Canada; (W.M.A.); (C.T.F.); (N.R.)
- Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
133
|
Suvanto M, Beesley J, Blomqvist C, Chenevix-Trench G, Khan S, Nevanlinna H. SNPs in lncRNA Regions and Breast Cancer Risk. Front Genet 2020; 11:550. [PMID: 32714364 PMCID: PMC7340126 DOI: 10.3389/fgene.2020.00550] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 05/07/2020] [Indexed: 01/18/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) play crucial roles in human physiology, and have been found to be associated with various cancers. Transcribed ultraconserved regions (T-UCRs) are a subgroup of lncRNAs conserved in several species, and are often located in cancer-related regions. Breast cancer is the most common cancer in women worldwide and the leading cause of female cancer deaths. We investigated the association of genetic variants in lncRNA and T-UCR regions with breast cancer risk to uncover candidate loci for further analysis. Our focus was on low-penetrance variants that can be discovered in a large dataset. We selected 565 regions of lncRNAs and T-UCRs that are expressed in breast or breast cancer tissue, or show expression correlation to major breast cancer associated genes. We studied the association of single nucleotide polymorphisms (SNPs) in these regions with breast cancer risk in the 122970 case samples and 105974 controls of the Breast Cancer Association Consortium's genome-wide data, and also by in silico functional analyses using Integrated Expression Quantitative trait and in silico prediction of GWAS targets (INQUISIT) and expression quantitative trait loci (eQTL) analysis. The eQTL analysis was carried out using the METABRIC dataset and analyses from GTEx and ncRNA eQTL databases. We found putative breast cancer risk variants (p < 1 × 10-5) targeting the lncRNA GABPB1-AS1 in INQUISIT and eQTL analysis. In addition, putative breast cancer risk associated SNPs (p < 1 × 10-5) in the region of two T-UCRs, uc.184 and uc.313, located in protein coding genes CPEB4 and TIAL1, respectively, targeted these genes in INQUISIT and in eQTL analysis. Other non-coding regions containing SNPs with the defined p-value and highly significant false discovery rate (FDR) for breast cancer risk association were discovered that may warrant further studies. These results suggest candidate lncRNA loci for further research on breast cancer risk and the molecular mechanisms.
Collapse
Affiliation(s)
- Maija Suvanto
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jonathan Beesley
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QL, Australia
| | - Carl Blomqvist
- Department of Oncology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Georgia Chenevix-Trench
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QL, Australia
| | - Sofia Khan
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
134
|
DNA Repair and Ovarian Carcinogenesis: Impact on Risk, Prognosis and Therapy Outcome. Cancers (Basel) 2020; 12:cancers12071713. [PMID: 32605254 PMCID: PMC7408288 DOI: 10.3390/cancers12071713] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 06/24/2020] [Indexed: 12/13/2022] Open
Abstract
There is ample evidence for the essential involvement of DNA repair and DNA damage response in the onset of solid malignancies, including ovarian cancer. Indeed, high-penetrance germline mutations in DNA repair genes are important players in familial cancers: BRCA1, BRCA2 mutations or mismatch repair, and polymerase deficiency in colorectal, breast, and ovarian cancers. Recently, some molecular hallmarks (e.g., TP53, KRAS, BRAF, RAD51C/D or PTEN mutations) of ovarian carcinomas were identified. The manuscript overviews the role of DNA repair machinery in ovarian cancer, its risk, prognosis, and therapy outcome. We have attempted to expose molecular hallmarks of ovarian cancer with a focus on DNA repair system and scrutinized genetic, epigenetic, functional, and protein alterations in individual DNA repair pathways (homologous recombination, non-homologous end-joining, DNA mismatch repair, base- and nucleotide-excision repair, and direct repair). We suggest that lack of knowledge particularly in non-homologous end joining repair pathway and the interplay between DNA repair pathways needs to be confronted. The most important genes of the DNA repair system are emphasized and their targeting in ovarian cancer will deserve further attention. The function of those genes, as well as the functional status of the entire DNA repair pathways, should be investigated in detail in the near future.
Collapse
|
135
|
Li H, Liu ZY, Wu N, Chen YC, Cheng Q, Wang J. PARP inhibitor resistance: the underlying mechanisms and clinical implications. Mol Cancer 2020; 19:107. [PMID: 32563252 PMCID: PMC7305609 DOI: 10.1186/s12943-020-01227-0] [Citation(s) in RCA: 239] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/11/2020] [Indexed: 12/31/2022] Open
Abstract
Due to the DNA repair defect, BRCA1/2 deficient tumor cells are more sensitive to PARP inhibitors (PARPi) through the mechanism of synthetic lethality. At present, several PAPRi targeting poly (ADP-ribose) polymerase (PARP) have been approved for ovarian cancer and breast cancer indications. However, PARPi resistance is ubiquitous in clinic. More than 40% BRCA1/2-deficient patients fail to respond to PARPi. In addition, lots of patients acquire PARPi resistance with prolonged oral administration of PARPi. Homologous recombination repair deficient (HRD), as an essential prerequisite of synthetic lethality, plays a vital role in killing tumor cells. Therefore, Homologous recombination repair restoration (HRR) becomes the predominant reason of PARPi resistance. Recently, it was reported that DNA replication fork protection also contributed to PARPi resistance in BRCA1/2-deficient cells and patients. Moreover, various factors, such as reversion mutations, epigenetic modification, restoration of ADP-ribosylation (PARylation) and pharmacological alteration lead to PARPi resistance as well. In this review, we reviewed the underlying mechanisms of PARP inhibitor resistance in detail and summarized the potential strategies to overcome PARPi resistance and increase PARPi sensitivity.
Collapse
Affiliation(s)
- He Li
- Hunan Clinical Research Center in Gynecologic Cancer, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283, Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Zhao-Yi Liu
- Hunan Clinical Research Center in Gynecologic Cancer, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283, Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Nayiyuan Wu
- Hunan Clinical Research Center in Gynecologic Cancer, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283, Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Yong-Chang Chen
- Hunan Clinical Research Center in Gynecologic Cancer, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283, Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Jing Wang
- Hunan Clinical Research Center in Gynecologic Cancer, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283, Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China. .,Department of Gynecologic Cancer, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283, Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China.
| |
Collapse
|
136
|
Bouwman P, van der Heijden I, van der Gulden H, de Bruijn R, Braspenning ME, Moghadasi S, Wessels LFA, Vreeswijk MPG, Jonkers J. Functional Categorization of BRCA1 Variants of Uncertain Clinical Significance in Homologous Recombination Repair Complementation Assays. Clin Cancer Res 2020; 26:4559-4568. [PMID: 32546644 DOI: 10.1158/1078-0432.ccr-20-0255] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/29/2020] [Accepted: 06/12/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Because BRCA1 is a high-risk breast/ovarian cancer susceptibility gene, BRCA1 sequence variants of uncertain clinical significance (VUS) complicate genetic counseling. As most VUS are rare, reliable classification based on clinical and genetic data is often impossible. However, all pathogenic BRCA1 variants analyzed result in defective homologous recombination DNA repair (HRR). Thus, BRCA1 VUS may be categorized based on their functional impact on this pathway. EXPERIMENTAL DESIGN Two hundred thirty-eight BRCA1 VUS-comprising most BRCA1 VUS known in the Netherlands and Belgium-were tested for their ability to complement Brca1-deficient mouse embryonic stem cells in HRR, using cisplatin and olaparib sensitivity assays and a direct repeat GFP (DR-GFP) HRR assay. Assays were validated using 25 known benign and 25 known pathogenic BRCA1 variants. For assessment of pathogenicity by a multifactorial likelihood analysis method, we collected clinical and genetic data for functionally deleterious VUS and VUS occurring in three or more families. RESULTS All three assays showed 100% sensitivity and specificity (95% confidence interval, 83%-100%). Out of 238 VUS, 45 showed functional defects, 26 of which were deleterious in all three assays. For 13 of these 26 variants, we could calculate the probability of pathogenicity using clinical and genetic data, resulting in the identification of 7 (likely) pathogenic variants. CONCLUSIONS We have functionally categorized 238 BRCA1 VUS using three different HRR-related assays. Classification based on clinical and genetic data alone for a subset of these variants confirmed the high sensitivity and specificity of our functional assays.
Collapse
Affiliation(s)
- Peter Bouwman
- Oncode Institute and Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Ingrid van der Heijden
- Oncode Institute and Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Hanneke van der Gulden
- Oncode Institute and Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Roebi de Bruijn
- Oncode Institute and Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,Oncode Institute and Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Merel E Braspenning
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Setareh Moghadasi
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Lodewyk F A Wessels
- Oncode Institute and Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Maaike P G Vreeswijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Jos Jonkers
- Oncode Institute and Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
137
|
Kaneyasu T, Mori S, Yamauchi H, Ohsumi S, Ohno S, Aoki D, Baba S, Kawano J, Miki Y, Matsumoto N, Nagasaki M, Yoshida R, Akashi-Tanaka S, Iwase T, Kitagawa D, Masuda K, Hirasawa A, Arai M, Takei J, Ide Y, Gotoh O, Yaguchi N, Nishi M, Kaneko K, Matsuyama Y, Okawa M, Suzuki M, Nezu A, Yokoyama S, Amino S, Inuzuka M, Noda T, Nakamura S. Prevalence of disease-causing genes in Japanese patients with BRCA1/2-wildtype hereditary breast and ovarian cancer syndrome. NPJ Breast Cancer 2020; 6:25. [PMID: 32566746 PMCID: PMC7293299 DOI: 10.1038/s41523-020-0163-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 04/30/2020] [Indexed: 12/30/2022] Open
Abstract
Panel sequencing of susceptibility genes for hereditary breast and ovarian cancer (HBOC) syndrome has uncovered numerous germline variants; however, their pathogenic relevance and ethnic diversity remain unclear. Here, we examined the prevalence of germline variants among 568 Japanese patients with BRCA1/2-wildtype HBOC syndrome and a strong family history. Pathogenic or likely pathogenic variants were identified on 12 causal genes for 37 cases (6.5%), with recurrence for 4 SNVs/indels and 1 CNV. Comparisons with non-cancer east-Asian populations and European familial breast cancer cohorts revealed significant enrichment of PALB2, BARD1, and BLM mutations. Younger onset was associated with but not predictive of these mutations. Significant somatic loss-of-function alterations were confirmed on the wildtype alleles of genes with germline mutations, including PALB2 additional somatic truncations. This study highlights Japanese-associated germline mutations among patients with BRCA1/2 wildtype HBOC syndrome and a strong family history, and provides evidence for the medical care of this high-risk population.
Collapse
Affiliation(s)
- Tomoko Kaneyasu
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku Tokyo, Japan
| | - Seiichi Mori
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku Tokyo, Japan
| | - Hideko Yamauchi
- Department of Breast Surgical Oncology, St. Luke’s International Hospital, 10-1 Akashi-cho, Chuo-ku Tokyo, Japan
| | - Shozo Ohsumi
- National Hospital Organization Shikoku Cancer Center, 160 Kou, Minamiumemoto-machi, Matsuyama, Ehime Japan
| | - Shinji Ohno
- Breast Oncology Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku Tokyo, Japan
| | - Daisuke Aoki
- Department of Obstetrics & Gynecology, Keio University School of Medicine, 35 Shinano-cho, Shinjuku-ku Tokyo, Japan
| | - Shinichi Baba
- Sagara Hospital, 3-31 Matsubara-cho, Kagoshima, Japan
| | - Junko Kawano
- Sagara Hospital, 3-31 Matsubara-cho, Kagoshima, Japan
| | - Yoshio Miki
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku Tokyo, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Fukuura 3-9, Kanazawa-ku Yokohama, Japan
| | - Masao Nagasaki
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi Japan
| | - Reiko Yoshida
- Department of Clinical Genetic Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku Tokyo, Japan
| | - Sadako Akashi-Tanaka
- Division of Breast Surgical Oncology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku Tokyo, Japan
| | - Takuji Iwase
- Breast Oncology Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku Tokyo, Japan
| | - Dai Kitagawa
- Breast Oncology Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku Tokyo, Japan
| | - Kenta Masuda
- Department of Obstetrics & Gynecology, Keio University School of Medicine, 35 Shinano-cho, Shinjuku-ku Tokyo, Japan
| | - Akira Hirasawa
- Department of Obstetrics & Gynecology, Keio University School of Medicine, 35 Shinano-cho, Shinjuku-ku Tokyo, Japan
| | - Masami Arai
- Department of Clinical Genetic Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku Tokyo, Japan
| | - Junko Takei
- Department of Breast Surgical Oncology, St. Luke’s International Hospital, 10-1 Akashi-cho, Chuo-ku Tokyo, Japan
| | - Yoshimi Ide
- Division of Breast Surgical Oncology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku Tokyo, Japan
| | - Osamu Gotoh
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku Tokyo, Japan
| | - Noriko Yaguchi
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku Tokyo, Japan
| | - Mitsuyo Nishi
- Sagara Hospital, 3-31 Matsubara-cho, Kagoshima, Japan
| | - Keika Kaneko
- National Hospital Organization Shikoku Cancer Center, 160 Kou, Minamiumemoto-machi, Matsuyama, Ehime Japan
| | - Yumi Matsuyama
- National Hospital Organization Shikoku Cancer Center, 160 Kou, Minamiumemoto-machi, Matsuyama, Ehime Japan
| | - Megumi Okawa
- Department of Breast Surgical Oncology, St. Luke’s International Hospital, 10-1 Akashi-cho, Chuo-ku Tokyo, Japan
| | - Misato Suzuki
- Department of Breast Surgical Oncology, St. Luke’s International Hospital, 10-1 Akashi-cho, Chuo-ku Tokyo, Japan
| | - Aya Nezu
- Breast Oncology Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku Tokyo, Japan
| | - Shiro Yokoyama
- Division of Breast Surgical Oncology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku Tokyo, Japan
| | - Sayuri Amino
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku Tokyo, Japan
| | - Mayuko Inuzuka
- Division of Breast Surgical Oncology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku Tokyo, Japan
| | - Tetsuo Noda
- Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku Tokyo, Japan
| | - Seigo Nakamura
- Division of Breast Surgical Oncology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku Tokyo, Japan
| |
Collapse
|
138
|
Kougioumtsidou N, Vavoulidis E, Nasioutziki M, Symeonidou M, Pratilas GC, Mareti E, Petousis S, Chatzikyriakidou A, Grimbizis G, Theodoridis T, Miliaras D, Dinas K, Zepiridis L. DNA methylation patterns of RAR-β2 and RASSF1A gene promoters in FNAB samples from Greek population with benign or malignant breast lesions. Diagn Cytopathol 2020; 49:153-164. [PMID: 32530576 DOI: 10.1002/dc.24513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/27/2020] [Accepted: 05/19/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Promoter hypermethylation is common in Breast Cancer (BC) with studies mainly in histological specimens showing frequent methylation of tumor suppressor genes (TSGs) compared with normal tissues. The aim of this study was to estimate the frequency of promoter methylation of RAR-β2 and RASSF1A genes in breast FNAB material aiming to evaluate the methylation status of these two genes as biomarker for detecting BC in Greek population. METHODS FNAB material from 104 patients was collected for cytological evaluation and epigenetic analysis. DNA was extracted and subjected to bisulfite conversion. A methylation-specific PCR was carried out and the final products were separated with electrophoresis in 2% agarose gels. RESULTS From 104 samples, RASSF1A hypermethylation was observed in 78 (75%) and RAR-β2 hypermethylation in 64 (61.6%). 84% and 78% of the cases diagnosed with breast malignancy (n = 50) were methylated for RASSF1A and RAR-β2, respectively. Methylated RASSF1A and RAR-β2 were also detected in 88.3% and 76.5% in samples diagnosed as suspicious for malignancy (n = 17) and in 57.2% of samples diagnosed with atypia (n = 14). The Odds Ratio for breast malignancy was 4.545 in patients with RASSF1A hypermethylation and 9.167 in patients with RAR-β2 hypermethylation underlying their promoter's methylation positive correlation with breast malignancy. CONCLUSION To optimize the sensitivity and specificity of this epigenetic setting, more TSGs related to BC should be gradually imported in our evaluated methylation panel and be validated in a larger study sample with the aim that the obtained epigenetic profiles will provide clinicians with valuable tools for management of BC patients in Greece.
Collapse
Affiliation(s)
- Niki Kougioumtsidou
- Faculty of Medicine, First Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Papageorgiou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Eleftherios Vavoulidis
- Faculty of Medicine, Second Department of Obstetrics and Gynaecology and Molecular Cytopathology Laboratory, Aristotle University of Thessaloniki, Hippokration General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Maria Nasioutziki
- Faculty of Medicine, Second Department of Obstetrics and Gynaecology and Molecular Cytopathology Laboratory, Aristotle University of Thessaloniki, Hippokration General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Marianthi Symeonidou
- Faculty of Medicine, First Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Papageorgiou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Georgios Chrysostomos Pratilas
- Faculty of Medicine, Second Department of Obstetrics and Gynaecology and Molecular Cytopathology Laboratory, Aristotle University of Thessaloniki, Hippokration General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Mareti
- Faculty of Medicine, Second Department of Obstetrics and Gynaecology and Molecular Cytopathology Laboratory, Aristotle University of Thessaloniki, Hippokration General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Stamatios Petousis
- Faculty of Medicine, Second Department of Obstetrics and Gynaecology and Molecular Cytopathology Laboratory, Aristotle University of Thessaloniki, Hippokration General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Anthoula Chatzikyriakidou
- Faculty of Medicine, Laboratory of Medical Biology-Genetics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Gregorios Grimbizis
- Faculty of Medicine, First Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Papageorgiou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Theodoros Theodoridis
- Faculty of Medicine, First Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Papageorgiou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Dimosthenis Miliaras
- Faculty of Medicine, First Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Papageorgiou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Konstantinos Dinas
- Faculty of Medicine, First Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Papageorgiou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Leonidas Zepiridis
- Faculty of Medicine, Second Department of Obstetrics and Gynaecology and Molecular Cytopathology Laboratory, Aristotle University of Thessaloniki, Hippokration General Hospital of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
139
|
Abstract
Recent breakthroughs in the fields of genomics and biology have resulted in a better understanding of diseases and their underlying biology. New targeted and immune-based therapies take advantage of these new discoveries to treat the patient individually. This scientific revolution toward personalized medicine reflects osteopathic medicine's emphasis on patient-centered care and its tenets, which go against the "one-size-fits-all" approach. The authors discuss the importance of applying osteopathic philosophy to the delivery of patient-directed cancer care revolutionized by scientific advances.
Collapse
|
140
|
Cervelli T, Lodovichi S, Bellè F, Galli A. Yeast-based assays for the functional characterization of cancer-associated variants of human DNA repair genes. MICROBIAL CELL 2020; 7:162-174. [PMID: 32656256 PMCID: PMC7328678 DOI: 10.15698/mic2020.07.721] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Technological advances are continuously revealing new genetic variants that are often difficult to interpret. As one of the most genetically tractable model organisms, yeast can have a central role in determining the consequences of human genetic variation. DNA repair gene mutations are associated with many types of cancers, therefore the evaluation of the functional impact of these mutations is crucial for risk assessment and for determining therapeutic strategies. Owing to the evolutionary conservation of DNA repair pathways between human cells and the yeast Saccharomyces cerevisiae, several functional assays have been developed. Here, we describe assays for variants of human genes belonging to the major DNA repair pathways divided in functional assays for human genes with yeast orthologues and human genes lacking a yeast orthologue. Human genes with orthologues can be studied by introducing the correspondent human mutations directly in the yeast gene or expressing the human gene carrying the mutations; while the only possible approach for human genes without a yeast orthologue is the heterologous expression. The common principle of these approaches is that the mutated gene determines a phenotypic alteration that can vary according to the gene studied and the domain of the protein. Here, we show how the versatility of yeast can help in classifying cancer-associated variants.
Collapse
Affiliation(s)
- Tiziana Cervelli
- Yeast Genetics and Genomics Group, Laboratory of Functional Genetics and Genomics, Institute of Clinical Physiology CNR, Via Moruzzi 1, 56125 Pisa, Italy
| | - Samuele Lodovichi
- Yeast Genetics and Genomics Group, Laboratory of Functional Genetics and Genomics, Institute of Clinical Physiology CNR, Via Moruzzi 1, 56125 Pisa, Italy
| | - Francesca Bellè
- Yeast Genetics and Genomics Group, Laboratory of Functional Genetics and Genomics, Institute of Clinical Physiology CNR, Via Moruzzi 1, 56125 Pisa, Italy
| | - Alvaro Galli
- Yeast Genetics and Genomics Group, Laboratory of Functional Genetics and Genomics, Institute of Clinical Physiology CNR, Via Moruzzi 1, 56125 Pisa, Italy
| |
Collapse
|
141
|
Tedaldi G, Tebaldi M, Zampiga V, Cangini I, Pirini F, Ferracci E, Danesi R, Arcangeli V, Ravegnani M, Martinelli G, Falcini F, Ulivi P, Calistri D. Male Breast Cancer: Results of the Application of Multigene Panel Testing to an Italian Cohort of Patients. Diagnostics (Basel) 2020; 10:E269. [PMID: 32365798 PMCID: PMC7277207 DOI: 10.3390/diagnostics10050269] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/21/2020] [Accepted: 04/28/2020] [Indexed: 12/24/2022] Open
Abstract
Male breast cancer (MBC) is a rare tumor, accounting for less than 1% of all breast cancers. In MBC, genetic predisposition plays an important role; however, only a few studies have investigated in depth the role of genes other than BRCA1 and BRCA2. We performed a Next-Generation Sequencing (NGS) analysis with a panel of 94 cancer predisposition genes on germline DNA from an Italian case series of 70 patients with MBC. Moreover, we searched for large deletions/duplications of BRCA1/2 genes through the Multiplex Ligation-dependent Probe Amplification (MLPA) technique. Through the combination of NGS and MLPA, we identified three pathogenic variants in the BRCA1 gene and six in the BRCA2 gene. Besides these alterations, we found six additional pathogenic/likely-pathogenic variants in PALB2, CHEK2, ATM, RAD51C, BAP1 and EGFR genes. From our study, BRCA1 and BRCA2 emerge as the main genes associated with MBC risk, but also other genes seem to be associated with the disease. Indeed, some of these genes have already been implicated in female breast cancer predisposition, but others are known to be involved in other types of cancer. Consequently, our results suggest that novel genes could be involved in MBC susceptibility, shedding new light on their role in cancer development.
Collapse
Affiliation(s)
- Gianluca Tedaldi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (V.Z.); (I.C.); (F.P.); (E.F.); (D.C.)
| | - Michela Tebaldi
- Biostatistics and Clinical Trials Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | - Valentina Zampiga
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (V.Z.); (I.C.); (F.P.); (E.F.); (D.C.)
| | - Ilaria Cangini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (V.Z.); (I.C.); (F.P.); (E.F.); (D.C.)
| | - Francesca Pirini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (V.Z.); (I.C.); (F.P.); (E.F.); (D.C.)
| | - Elisa Ferracci
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (V.Z.); (I.C.); (F.P.); (E.F.); (D.C.)
| | - Rita Danesi
- Romagna Cancer Registry, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (R.D.); (M.R.); (F.F.)
| | | | - Mila Ravegnani
- Romagna Cancer Registry, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (R.D.); (M.R.); (F.F.)
| | - Giovanni Martinelli
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | - Fabio Falcini
- Romagna Cancer Registry, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (R.D.); (M.R.); (F.F.)
| | - Paola Ulivi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (V.Z.); (I.C.); (F.P.); (E.F.); (D.C.)
| | - Daniele Calistri
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (V.Z.); (I.C.); (F.P.); (E.F.); (D.C.)
| |
Collapse
|
142
|
Behl S, Hamel N, de Ladurantaye M, Lepage S, Lapointe R, Mes-Masson AM, Foulkes WD. Founder BRCA1/BRCA2/PALB2 pathogenic variants in French-Canadian breast cancer cases and controls. Sci Rep 2020; 10:6491. [PMID: 32300229 PMCID: PMC7162921 DOI: 10.1038/s41598-020-63100-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 03/21/2020] [Indexed: 11/08/2022] Open
Abstract
Inherited germline pathogenic variants are responsible for ~5% of breast cancer globally. Through rapid expansion and isolation since immigration in the early 17th century, French Canadians are a relatively genetically homogenous founder population and therefore represent a unique demographic for genetic contributions to disease. To date, twenty variants in BRCA1, BRCA2, and PALB2 that predispose families to breast and ovarian cancer have been identified as recurring in the French-Canadian founder population. Our objective was to evaluate the clinical efficacy and validity of targeted genetic testing for these variants in Montreal French Canadians. A total of 555 breast cancer cases unselected for family history or age of diagnosis were genotyped, along with 1940 controls without a personal or family history of cancer. A Sequenom genotyping assay identified a pathogenic variant in 0.2% (5 of 1940) of cancer-free controls, and 3.8% (21/555) of breast cancer cases. Almost 10% (12/113) of early onset cases were heterozygous for founder BRCA1 or BRCA2 pathogenic variant. Of twenty variants tested, only seven were identified in this study. The option of providing this test as population-based screening is discussed.
Collapse
Affiliation(s)
- Supriya Behl
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Nancy Hamel
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Manon de Ladurantaye
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Stéphanie Lepage
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
- Institut du cancer de Montréal, Montréal, Québec, Canada
| | - Réjean Lapointe
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
- Institut du cancer de Montréal, Montréal, Québec, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - William D Foulkes
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada.
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
- Department of Medical Genetics, Jewish General Hospital, Montreal, Quebec, Canada.
| |
Collapse
|
143
|
53BP1 Accumulation in Circulating Tumor Cells Identifies Chemotherapy-Responsive Metastatic Breast Cancer Patients. Cancers (Basel) 2020; 12:cancers12040930. [PMID: 32283863 PMCID: PMC7226269 DOI: 10.3390/cancers12040930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/03/2020] [Accepted: 04/07/2020] [Indexed: 12/17/2022] Open
Abstract
Evidence suggests that the DNA end-binding protein p53-binding protein 1 (53BP1) is down-regulated in subsets of breast cancer. Circulating tumor cells (CTCs) provide accessible “biopsy material” to track cell traits and functions and their alterations during treatment. Here, we prospectively monitored the 53BP1 status in CTCs from 67 metastatic breast cancer (MBC) patients with HER2- CTCs and known hormone receptor (HR) status of the primary tumor and/or metastases before, during, and at the end of chemotherapeutic treatment with Eribulin. Nuclear 53BP1 staining and genomic integrity were evaluated by immunocytochemical and whole-genome-amplification-based polymerase chain reaction (PCR) analysis, respectively. Comparative analysis of CTCs from patients with triple-negative and HR+ tumors revealed elevated 53BP1 levels in CTCs from patients with HR+ metastases, particularly following chemotherapeutic treatment. Differences in nuclear 53BP1 signals did not correlate with genomic integrity in CTCs at baseline or with nuclear γH2AX signals in MBC cell lines, indicating that 53BP1 detected features beyond DNA damage. Kaplan–Meier analysis revealed an increasing association between nuclear 53BP1-positivity and progression-free survival (PFS) during chemotherapy until the final visit. Our data suggest that 53BP1 detection in CTCs could be a useful marker to capture dynamic changes of chemotherapeutic responsiveness in triple-negative and HR+ MBC.
Collapse
|
144
|
Oxidative Damage in Sporadic Colorectal Cancer: Molecular Mapping of Base Excision Repair Glycosylases in Colorectal Cancer Patients. Int J Mol Sci 2020; 21:ijms21072473. [PMID: 32252452 PMCID: PMC7177219 DOI: 10.3390/ijms21072473] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress with subsequent premutagenic oxidative DNA damage has been implicated in colorectal carcinogenesis. The repair of oxidative DNA damage is initiated by lesion-specific DNA glycosylases (hOGG1, NTH1, MUTYH). The direct evidence of the role of oxidative DNA damage and its repair is proven by hereditary syndromes (MUTYH-associated polyposis, NTHL1-associated tumor syndrome), where germline mutations cause loss-of-function in glycosylases of base excision repair, thus enabling the accumulation of oxidative DNA damage and leading to the adenoma-colorectal cancer transition. Unrepaired oxidative DNA damage often results in G:C>T:A mutations in tumor suppressor genes and proto-oncogenes and widespread occurrence of chromosomal copy-neutral loss of heterozygosity. However, the situation is more complicated in complex and heterogeneous disease, such as sporadic colorectal cancer. Here we summarized our current knowledge of the role of oxidative DNA damage and its repair on the onset, prognosis and treatment of sporadic colorectal cancer. Molecular and histological tumor heterogeneity was considered. Our study has also suggested an additional important source of oxidative DNA damage due to intestinal dysbiosis. The roles of base excision repair glycosylases (hOGG1, MUTYH) in tumor and adjacent mucosa tissues of colorectal cancer patients, particularly in the interplay with other factors (especially microenvironment), deserve further attention. Base excision repair characteristics determined in colorectal cancer tissues reflect, rather, a disease prognosis. Finally, we discuss the role of DNA repair in the treatment of colon cancer, since acquired or inherited defects in DNA repair pathways can be effectively used in therapy.
Collapse
|
145
|
Hatano Y, Tamada M, Matsuo M, Hara A. Molecular Trajectory of BRCA1 and BRCA2 Mutations. Front Oncol 2020; 10:361. [PMID: 32269964 PMCID: PMC7109296 DOI: 10.3389/fonc.2020.00361] [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: 12/06/2019] [Accepted: 03/02/2020] [Indexed: 12/12/2022] Open
Abstract
Every cancer carries genomic mutations. Although almost all these mutations arise after fertilization, a minimal count of cancer predisposition mutations are already present at the time of genesis of germ cells. Of the cancer predisposition genes identified to date, BRCA1 and BRCA2 have been determined to be associated with hereditary breast and ovarian cancer syndrome. Such cancer predisposition genes have recently been attracting attention owing to the emergence of molecular genetics, thus, affecting the strategy of cancer prevention, diagnostics, and therapeutics. In this review, we summarize the molecular significance of these two BRCA genes. First, we provide a brief history of BRCA1 and BRCA2, including their identification as cancer predisposition genes and recognition as members in the Fanconi anemia pathway. Next, we describe the molecular function and interaction of BRCA proteins, and thereafter, describe the patterns of BRCA dysfunction. Subsequently, we present emerging evidence on mutational signatures to determine the effects of BRCA disorders on the mutational process in cancer cells. Currently, BRCA genes serve as principal targets for clinical molecular oncology, be they germline or sporadic mutations. Moreover, comprehensive cancer genome analyses enable us to not only recognize the current status of the known cancer driver gene mutations but also divulge the past mutational processes and predict the future biological behavior of cancer through the molecular trajectory of genomic alterations.
Collapse
Affiliation(s)
- Yuichiro Hatano
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | | | | | | |
Collapse
|
146
|
Jia G, Lu Y, Wen W, Long J, Liu Y, Tao R, Li B, Denny JC, Shu XO, Zheng W. Evaluating the Utility of Polygenic Risk Scores in Identifying High-Risk Individuals for Eight Common Cancers. JNCI Cancer Spectr 2020; 4:pkaa021. [PMID: 32596635 PMCID: PMC7306192 DOI: 10.1093/jncics/pkaa021] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 01/14/2020] [Accepted: 03/06/2020] [Indexed: 12/13/2022] Open
Abstract
Background Genome-wide association studies have identified common genetic risk variants in many loci associated with multiple cancers. We sought to systematically evaluate the utility of these risk variants in identifying high-risk individuals for eight common cancers. Methods We constructed polygenic risk scores (PRS) using genome-wide association studies–identified risk variants for each cancer. Using data from 400 812 participants of European descent in a population-based cohort study, UK Biobank, we estimated hazard ratios associated with PRS using Cox proportional hazard models and evaluated the performance of the PRS in cancer risk prediction and their ability to identify individuals at more than a twofold elevated risk, a risk level comparable to a moderate-penetrance mutation in known cancer predisposition genes. Results During a median follow-up of 5.8 years, 14 584 incident case patients of cancers were identified (ranging from 358 epithelial ovarian cancer case patients to 4430 prostate cancer case patients). Compared with those at an average risk, individuals among the highest 5% of the PRS had a two- to threefold elevated risk for cancer of the prostate, breast, pancreas, colorectal, or ovary, and an approximately 1.5-fold elevated risk of cancer of the lung, bladder, or kidney. The areas under the curve ranged from 0.567 to 0.662. Using PRS, 40.4% of the study participants can be classified as having more than a twofold elevated risk for at least one site-specific cancer. Conclusions A large proportion of the general population can be identified at an elevated cancer risk by PRS, supporting the potential clinical utility of PRS for personalized cancer risk prediction.
Collapse
Affiliation(s)
- Guochong Jia
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yingchang Lu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ying Liu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ran Tao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bingshan Li
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
147
|
Variants of uncertain significance in the era of high-throughput genome sequencing: a lesson from breast and ovary cancers. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:46. [PMID: 32127026 PMCID: PMC7055088 DOI: 10.1186/s13046-020-01554-6] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 02/26/2020] [Indexed: 02/06/2023]
Abstract
The promising expectations about personalized medicine have opened the path to routine large-scale sequencing and increased the importance of genetic counseling for hereditary cancers, among which hereditary breast and ovary cancers (HBOC) have a major impact. High-throughput sequencing, or Next-Generation Sequencing (NGS), has improved cancer patient management, ameliorating diagnosis and treatment decisions. In addition to its undeniable clinical utility, NGS is also unveiling a large number of variants that we are still not able to clearly define and classify, the variants of uncertain significance (VUS), which account for about 40% of total variants. At present, VUS use in the clinical context is challenging. Medical reports may omit this kind of data and, even when included, they limit the clinical utility of genetic information. This has prompted the scientific community to seek easily applicable tests to accurately classify VUS and increase the amount of usable information from NGS data. In this review, we will focus on NGS and classification systems for VUS investigation, with particular attention on HBOC-related genes and in vitro functional tests developed for ameliorating and accelerating variant classification in cancer.
Collapse
|
148
|
Chung SH, Woldenberg N, Roth AR, Masamed R, Conlon W, Cohen JG, Joines MM, Patel MK. BRCA and Beyond: Comprehensive Image-rich Review of Hereditary Breast and Gynecologic Cancer Syndromes. Radiographics 2020; 40:306-325. [DOI: 10.1148/rg.2020190084] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Stephanie Histed Chung
- From the Departments of Radiology (S.H.C., R.M., M.M.J., M.K.P.), Clinical Genetics (W.C.), and Obstetrics and Gynecology (J.G.C.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, Calif; Hoag Hospital, Newport Harbor Radiology Associates, Newport Beach, Calif (N.W.); and Department of Radiology, Olive View–UCLA Medical Center, Sylmar, Calif (A.R.R.)
| | - Nina Woldenberg
- From the Departments of Radiology (S.H.C., R.M., M.M.J., M.K.P.), Clinical Genetics (W.C.), and Obstetrics and Gynecology (J.G.C.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, Calif; Hoag Hospital, Newport Harbor Radiology Associates, Newport Beach, Calif (N.W.); and Department of Radiology, Olive View–UCLA Medical Center, Sylmar, Calif (A.R.R.)
| | - Antoinette R. Roth
- From the Departments of Radiology (S.H.C., R.M., M.M.J., M.K.P.), Clinical Genetics (W.C.), and Obstetrics and Gynecology (J.G.C.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, Calif; Hoag Hospital, Newport Harbor Radiology Associates, Newport Beach, Calif (N.W.); and Department of Radiology, Olive View–UCLA Medical Center, Sylmar, Calif (A.R.R.)
| | - Rinat Masamed
- From the Departments of Radiology (S.H.C., R.M., M.M.J., M.K.P.), Clinical Genetics (W.C.), and Obstetrics and Gynecology (J.G.C.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, Calif; Hoag Hospital, Newport Harbor Radiology Associates, Newport Beach, Calif (N.W.); and Department of Radiology, Olive View–UCLA Medical Center, Sylmar, Calif (A.R.R.)
| | - Wendy Conlon
- From the Departments of Radiology (S.H.C., R.M., M.M.J., M.K.P.), Clinical Genetics (W.C.), and Obstetrics and Gynecology (J.G.C.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, Calif; Hoag Hospital, Newport Harbor Radiology Associates, Newport Beach, Calif (N.W.); and Department of Radiology, Olive View–UCLA Medical Center, Sylmar, Calif (A.R.R.)
| | - Joshua G. Cohen
- From the Departments of Radiology (S.H.C., R.M., M.M.J., M.K.P.), Clinical Genetics (W.C.), and Obstetrics and Gynecology (J.G.C.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, Calif; Hoag Hospital, Newport Harbor Radiology Associates, Newport Beach, Calif (N.W.); and Department of Radiology, Olive View–UCLA Medical Center, Sylmar, Calif (A.R.R.)
| | - Melissa M. Joines
- From the Departments of Radiology (S.H.C., R.M., M.M.J., M.K.P.), Clinical Genetics (W.C.), and Obstetrics and Gynecology (J.G.C.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, Calif; Hoag Hospital, Newport Harbor Radiology Associates, Newport Beach, Calif (N.W.); and Department of Radiology, Olive View–UCLA Medical Center, Sylmar, Calif (A.R.R.)
| | - Maitraya K. Patel
- From the Departments of Radiology (S.H.C., R.M., M.M.J., M.K.P.), Clinical Genetics (W.C.), and Obstetrics and Gynecology (J.G.C.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, Calif; Hoag Hospital, Newport Harbor Radiology Associates, Newport Beach, Calif (N.W.); and Department of Radiology, Olive View–UCLA Medical Center, Sylmar, Calif (A.R.R.)
| |
Collapse
|
149
|
Otsuka I, Matsuura T. Screening and Prevention for High-Grade Serous Carcinoma of the Ovary Based on Carcinogenesis-Fallopian Tube- and Ovarian-Derived Tumors and Incessant Retrograde Bleeding. Diagnostics (Basel) 2020; 10:E120. [PMID: 32098383 PMCID: PMC7168061 DOI: 10.3390/diagnostics10020120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 02/06/2023] Open
Abstract
High-grade serous carcinoma (HGSC) is the most common and lethal subtype of ovarian carcinoma. Many HGSCs are now believed to originate in the fallopian tube epithelium; ovarian surface epithelium is another possible origin. Thus, current screening methods, i.e., ultrasonography and serum CA-125 measurements, have a limitation in their early detection. Recently, circulating biomarkers, such as tumor DNA, autoantibody, and microRNA, have been investigated to detect HGSCs. As cancer cells in the fallopian tube flow into the endometrial cavity, the detection of exfoliated cells, tumor DNA, and proteome from samples obtained from the endometrial cavity or the cervix may be useful. The risk of ovarian serous carcinoma is affected by the use of oral contraceptive and menopausal hormone therapy (MHT). MHT regimens causing endometrial bleeding increase serous carcinoma risk, hence, incessant retrograde bleeding from the endometrial cavity into the Douglas pouch appears to play an important role in high-grade serous carcinogenesis. In this review, we provide an overview of current and novel screening methods and prevention approaches for ovarian and fallopian tube HGSC.
Collapse
Affiliation(s)
- Isao Otsuka
- Department of Obstetrics and Gynecology, Kameda Medical Center, Kamogawa 296-8602, Japan;
| | | |
Collapse
|
150
|
Angeli D, Salvi S, Tedaldi G. Genetic Predisposition to Breast and Ovarian Cancers: How Many and Which Genes to Test? Int J Mol Sci 2020; 21:E1128. [PMID: 32046255 PMCID: PMC7038038 DOI: 10.3390/ijms21031128] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 12/19/2022] Open
Abstract
Breast and ovarian cancers are some of the most common tumors in females, and the genetic predisposition is emerging as one of the key risk factors in the development of these two malignancies. BRCA1 and BRCA2 are the best-known genes associated with hereditary breast and ovarian cancer. However, recent advances in molecular techniques, Next-Generation Sequencing in particular, have led to the identification of many new genes involved in the predisposition to breast and/or ovarian cancer, with different penetrance estimates. TP53, PTEN, STK11, and CDH1 have been identified as high penetrance genes for the risk of breast/ovarian cancers. Besides them, PALB2, BRIP1, ATM, CHEK2, BARD1, NBN, NF1, RAD51C, RAD51D and mismatch repair genes have been recognized as moderate and low penetrance genes, along with other genes encoding proteins involved in the same pathways, possibly associated with breast/ovarian cancer risk. In this review, we summarize the past and more recent findings in the field of cancer predisposition genes, with insights into the role of the encoded proteins and the associated genetic disorders. Furthermore, we discuss the possible clinical utility of genetic testing in terms of prevention protocols and therapeutic approaches.
Collapse
Affiliation(s)
- Davide Angeli
- Biostatistics and Clinical Trials Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | - Samanta Salvi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | - Gianluca Tedaldi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| |
Collapse
|