1
|
Minello A, Carreira A. BRCA1/2 Haploinsufficiency: Exploring the Impact of Losing one Allele. J Mol Biol 2024; 436:168277. [PMID: 37714298 DOI: 10.1016/j.jmb.2023.168277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023]
Abstract
Since their discovery in the late 20th century, significant progress has been made in elucidating the functions of the tumor suppressor proteins BRCA1 and BRCA2. These proteins play vital roles in maintaining genome integrity, including DNA repair, replication fork protection, and chromosome maintenance. It is well-established that germline mutations in BRCA1 and BRCA2 increase the risk of breast and ovarian cancer; however, the precise mechanism underlying tumor formation in this context is not fully understood. Contrary to the long-standing belief that the loss of the second wild-type allele is necessary for tumor development, a growing body of evidence suggests that tumorigenesis can occur despite the presence of a single functional allele. This entails that heterozygosity in BRCA1/2 confers haploinsufficiency, where a single copy of the gene is not sufficient to fully suppress tumor formation. Here we provide an overview of the findings and the ongoing debate regarding BRCA haploinsufficiency. We further put out the challenges in studying this topic and discuss its potential relevance in the prevention and treatment of BRCA-related cancers.
Collapse
Affiliation(s)
- Anna Minello
- Institut Curie, PSL Research University, CNRS, UMR3348, F-91405 Orsay, France; Paris-Saclay University CNRS, UMR3348, F-91405 Orsay, France
| | - Aura Carreira
- Institut Curie, PSL Research University, CNRS, UMR3348, F-91405 Orsay, France; Paris-Saclay University CNRS, UMR3348, F-91405 Orsay, France; Genome Instability and Cancer Predisposition Lab, Department of Genome Dynamics and Function, Centro de Biologia Molecular Severo Ochoa (CBMSO, CSIC-UAM), Madrid 28049, Spain.
| |
Collapse
|
2
|
Korneenko TV, Pestov NB. Oncogenic BRCA1,2 Mutations in the Human Lineage-A By-Product of Sexual Selection? Biomedicines 2023; 12:22. [PMID: 38275383 PMCID: PMC10813183 DOI: 10.3390/biomedicines12010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 01/27/2024] Open
Abstract
In this review, we discuss the long-known problem of tissue-specific carcinogenesis in BRCA1 and BRCA2 mutation carriers: while the genes are expressed ubiquitously, increased cancer risk is observed mostly in the breast and ovaries, and to a much lesser extent, in some other tissues such as the prostate or pancreas. We reevaluate hypotheses on the evolutionary origin of these mutations in humans. Also, we align together the reports that at least some great apes have much lower risks of epithelial cancers in general and breast cancer in particular with the fact that humans have more voluminous breast tissue as compared to their closest extant relatives, particularly chimpanzees and bonobos. We conjecture that this disparity may be a consequence of sexual selection, augmented via selection for enhanced lactation. Further, we argue that there is an organ-specific enigma similar to the Peto paradox: breast cancer risk in humans is only minimally correlated with breast size. These considerations lead to the hypothesis that, along with the evolutionary development of larger breasts in humans, additional changes have played a balancing role in suppressing breast cancer. These yet-to-be-discovered mechanisms, while purely speculative, may be valuable to understanding human breast cancer, though they may not be exclusive to the mammary gland epithelial cells. Combining these themes, we review some anti-carcinogenesis preventive strategies and prospects of new interventions against breast cancer.
Collapse
Affiliation(s)
- Tatyana V. Korneenko
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia
| | - Nikolay B. Pestov
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia
- Institute of Biomedical Chemistry, Moscow 119121, Russia
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Moscow 108819, Russia
| |
Collapse
|
3
|
Oubaddou Y, Ben Ali F, Oubaqui FE, Qmichou Z, Bakri Y, Rabii Ameziane RA. The Tumor Suppressor BRCA1/2, Cancer Susceptibility and Genome Instability in Gynecological and Mammary Cancers. Asian Pac J Cancer Prev 2023; 24:3139-3153. [PMID: 37774066 PMCID: PMC10762740 DOI: 10.31557/apjcp.2023.24.9.3139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/10/2023] [Indexed: 10/01/2023] Open
Abstract
BRCA1 and BRCA2 germline alterations highly predispose women to breast and ovarian cancers. They are mostly found within the TNBC (Triple-Negative Breast Cancer) and the HGSOC (High-Grade Serous Ovarian Carcinoma) subsets, known by an aggressive phenotype, the lack of therapeutic targets and poor prognosis. Importantly, there is an increased risk for cervical cancer in BRCA1 and BRCA2 mutation carriers that raises questions about the link between the HPV-driven genome instability and BRCA1 and BRCA2 germline mutations. Clinical, preclinical, and in vitro studies explained the increased risk for breast and ovarian cancers by genome instability resulting from the lack or loss of many functions related to BRCA1 or BRCA2 proteins such as DNA damage repair, stalled forks and R-loops resolution, transcription regulation, cell cycle control, and oxidative stress. In this review, we decipher the relationship between BRCA1/2 alterations and genomic instability leading to gynecomammary cancers through results from patients, mice, and cell lines. Understanding the early events of BRCA1/2-driven genomic instability in gynecomammary cancers would help to find new biomarkers for early diagnosis, improve the sensitivity of emerging therapies such as PARP inhibitors, and reveal new potential therapeutic targets.
Collapse
Affiliation(s)
- Yassire Oubaddou
- Laboratory of Biology of Human Pathologies (BioPatH), Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco.
| | - Fatima Ben Ali
- Laboratory of Biology of Human Pathologies (BioPatH), Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco.
| | - Fatima Ezzahrae Oubaqui
- Laboratory of Biology of Human Pathologies (BioPatH), Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco.
- Medical Biotechnology Center, Moroccan Foundation for Advanced Science, Innovation and Research (MAScIR), Rabat, Morocco.
| | - Zineb Qmichou
- Medical Biotechnology Center, Moroccan Foundation for Advanced Science, Innovation and Research (MAScIR), Rabat, Morocco.
| | - Youssef Bakri
- Laboratory of Biology of Human Pathologies (BioPatH), Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco.
| | - Rabii Ameziane Rabii Ameziane
- Laboratory of Biology of Human Pathologies (BioPatH), Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco.
| |
Collapse
|
4
|
Prochownik EV, Wang H. Normal and Neoplastic Growth Suppression by the Extended Myc Network. Cells 2022; 11:747. [PMID: 35203395 PMCID: PMC8870482 DOI: 10.3390/cells11040747] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/09/2022] [Accepted: 02/15/2022] [Indexed: 12/20/2022] Open
Abstract
Among the first discovered and most prominent cellular oncogenes is MYC, which encodes a bHLH-ZIP transcription factor (Myc) that both activates and suppresses numerous genes involved in proliferation, energy production, metabolism and translation. Myc belongs to a small group of bHLH-ZIP transcriptional regulators (the Myc Network) that includes its obligate heterodimerization partner Max and six "Mxd proteins" (Mxd1-4, Mnt and Mga), each of which heterodimerizes with Max and largely opposes Myc's functions. More recently, a second group of bHLH-ZIP proteins (the Mlx Network) has emerged that bears many parallels with the Myc Network. It is comprised of the Myc-like factors ChREBP and MondoA, which, in association with the Max-like member Mlx, regulate smaller and more functionally restricted repertoires of target genes, some of which are shared with Myc. Opposing ChREBP and MondoA are heterodimers comprised of Mlx and Mxd1, Mxd4 and Mnt, which also structurally and operationally link the two Networks. We discuss here the functions of these "Extended Myc Network" members, with particular emphasis on their roles in suppressing normal and neoplastic growth. These roles are complex due to the temporal- and tissue-restricted expression of Extended Myc Network proteins in normal cells, their regulation of both common and unique target genes and, in some cases, their functional redundancy.
Collapse
Affiliation(s)
- Edward V. Prochownik
- Division of Hematology/Oncology, The Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA;
- The Department of Microbiology and Molecular Genetics, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
- The Hillman Cancer Center of UPMC, Pittsburgh, PA 15224, USA
- The Pittsburgh Liver Research Center, Pittsburgh, PA 15224, USA
| | - Huabo Wang
- Division of Hematology/Oncology, The Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA;
| |
Collapse
|
5
|
Li N, Zethoven M, McInerny S, Devereux L, Huang YK, Thio N, Cheasley D, Gutiérrez-Enríquez S, Moles-Fernández A, Diez O, Nguyen-Dumont T, Southey MC, Hopper JL, Simard J, Dumont M, Soucy P, Meindl A, Schmutzler R, Schmidt MK, Adank MA, Andrulis IL, Hahnen E, Engel C, Lesueur F, Girard E, Neuhausen SL, Ziv E, Allen J, Easton DF, Scott RJ, Gorringe KL, James PA, Campbell IG. Evaluation of the association of heterozygous germline variants in NTHL1 with breast cancer predisposition: an international multi-center study of 47,180 subjects. NPJ Breast Cancer 2021; 7:52. [PMID: 33980861 PMCID: PMC8115524 DOI: 10.1038/s41523-021-00255-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
Bi-allelic loss-of-function (LoF) variants in the base excision repair (BER) gene NTHL1 cause a high-risk hereditary multi-tumor syndrome that includes breast cancer, but the contribution of heterozygous variants to hereditary breast cancer is unknown. An analysis of 4985 women with breast cancer, enriched for familial features, and 4786 cancer-free women revealed significant enrichment for NTHL1 LoF variants. Immunohistochemistry confirmed reduced NTHL1 expression in tumors from heterozygous carriers but the NTHL1 bi-allelic loss characteristic mutational signature (SBS 30) was not present. The analysis was extended to 27,421 breast cancer cases and 19,759 controls from 10 international studies revealing 138 cases and 93 controls with a heterozygous LoF variant (OR 1.06, 95% CI: 0.82-1.39) and 316 cases and 179 controls with a missense variant (OR 1.31, 95% CI: 1.09-1.57). Missense variants selected for deleterious features by a number of in silico bioinformatic prediction tools or located within the endonuclease III functional domain showed a stronger association with breast cancer. Somatic sequencing of breast cancers from carriers indicated that the risk associated with NTHL1 appears to operate through haploinsufficiency, consistent with other described low-penetrance breast cancer genes. Data from this very large international multicenter study suggests that heterozygous pathogenic germline coding variants in NTHL1 may be associated with low- to moderate- increased risk of breast cancer.
Collapse
Affiliation(s)
- Na Li
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Vic, Australia
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Vic, Australia
| | - Magnus Zethoven
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Bioinformatics Core Facility, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Simone McInerny
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Vic, Australia
| | - Lisa Devereux
- Lifepool, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Yu-Kuan Huang
- Upper Gastrointestinal Translational Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Melbourne, Vic, Australia
| | - Niko Thio
- Bioinformatics Core Facility, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Dane Cheasley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Vic, Australia
| | - Sara Gutiérrez-Enríquez
- Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO); Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Alejandro Moles-Fernández
- Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO); Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Orland Diez
- Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO); Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Area of Clinical and Molecular Genetics, Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Tu Nguyen-Dumont
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - Melissa C Southey
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jacques Simard
- Genomics Center, Centre Hospitalier Universitaire de Québec - Université Laval Research Center, Quebec, Canada
| | - Martine Dumont
- Genomics Center, Centre Hospitalier Universitaire de Québec - Université Laval Research Center, Quebec, Canada
| | - Penny Soucy
- Genomics Center, Centre Hospitalier Universitaire de Québec - Université Laval Research Center, Quebec, Canada
| | - Alfons Meindl
- University of Munich, Campus Großhadern, Department of Gynecology and Obstetrics, Munich, Germany
| | - Rita Schmutzler
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Familial Breast and Ovarian Cancer, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Integrated Oncology (CIO), Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
| | - Marjanka K Schmidt
- Division of Molecular Pathology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek hospital, Amsterdam, The Netherlands
| | - Muriel A Adank
- Family Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Irene L Andrulis
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Eric Hahnen
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Familial Breast and Ovarian Cancer, Cologne, Germany
| | - Christoph Engel
- Leipzig Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Fabienne Lesueur
- Inserm, U900, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | - Elodie Girard
- Inserm, U900, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | - Susan L Neuhausen
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Elad Ziv
- Department of Medicine, University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Jamie Allen
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Discipline of Medical Genetics, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- Division of Molecular Medicine, Pathology North, Newcastle, NSW, Australia
| | - Kylie L Gorringe
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Vic, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
- Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Paul A James
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Vic, Australia
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Vic, Australia
| | - Ian G Campbell
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Vic, Australia.
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
6
|
BECN1 and BRCA1 Deficiency Sensitizes Ovarian Cancer to Platinum Therapy and Confers Better Prognosis. Biomedicines 2021; 9:biomedicines9020207. [PMID: 33670664 PMCID: PMC7922320 DOI: 10.3390/biomedicines9020207] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/21/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022] Open
Abstract
Background: BRCA1, BECN1 and TP53 are three tumor suppressor genes located on chromosome 17 and frequently found deleted, silenced, or mutated in many cancers. These genes are involved in autophagy, apoptosis, and drug resistance in ovarian cancer. Haploinsufficiency or loss-of-function of either TP53, BRCA1 or BECN1 correlates with enhanced predisposition to cancer development and progression, and chemoresistance. Expectedly, the combined altered expression of these three tumor suppressor genes worsens the prognosis of ovarian cancer patients. However, whether such a genotypic pattern indeed affects the chemo-responsiveness to standard chemotherapy thus worsening patients’ survival has not been validated in a large cohort of ovarian cancer patients. Aim: We interrogated datasets from the TCGA database to analyze how the expression of these three tumor suppressor genes impacts on the clinical response to platinum-based chemotherapy thus affecting the survival of ovarian cancer patients. Results and conclusion: Compared to EOC with homozygous expression of BECN1 and BRCA1, tumors expressing low mRNA expression of these two tumor suppressor genes (either because of shallow (monoallelic) co-deletion or of promoter hypermethylation), showed higher sensitivity to platinum-based therapies and were associated with a better prognosis of ovarian cancer-bearing patients. This outcome was independent of TP53 status, though it was statistically more significant in the cohort of patients with mutated TP53. Thus, sensitivity to platinum therapy (and probably to other chemotherapeutics) correlates with low expression of a combination of critical tumor suppressor genes. Our study highlights the importance of thoroughly assessing the genetic lesions of the most frequently mutated genes to stratify the patients in view of a personalized therapy. More importantly, the present findings suggest that targeting the function of both BECN1 and BRCA1 could be a strategy to restore chemosensitivity in refractory tumors.
Collapse
|
7
|
Human mutational constraint as a tool to understand biology of rare and emerging bone marrow failure syndromes. Blood Adv 2020; 4:5232-5245. [PMID: 33104793 DOI: 10.1182/bloodadvances.2020002687] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/16/2020] [Indexed: 12/17/2022] Open
Abstract
Inherited bone marrow failure (IBMF) syndromes are rare blood disorders characterized by hematopoietic cell dysfunction and predisposition to hematologic malignancies. Despite advances in the understanding of molecular pathogenesis of these heterogeneous diseases, genetic variant interpretation, genotype-phenotype correlation, and outcome prognostication remain difficult. As new IBMF and other myelodysplastic syndrome (MDS) predisposition genes continue to be discovered (frequently in small kindred studies), there is an increasing need for a systematic framework to evaluate penetrance and prevalence of mutations in genes associated with IBMF phenotypes. To address this need, we analyzed population-based genomic data from >125 000 individuals in the Genome Aggregation Database for loss-of-function (LoF) variants in 100 genes associated with IBMF. LoF variants in genes associated with IBMF/MDS were present in 0.426% of individuals. Heterozygous LoF variants in genes in which haploinsufficiency is associated with IBMF/MDS were identified in 0.422% of the population; homozygous LoF variants associated with autosomal recessive IBMF/MDS diseases were identified in only .004% of the cohort. Using age distribution of LoF variants and 2 measures of mutational constraint, LOEUF ("loss-of-function observed/expected upper bound fraction") and pLI ("probability of being loss-of-function intolerance"), we evaluated the pathogenicity, tolerance, and age-related penetrance of LoF mutations in specific genes associated with IBMF syndromes. This analysis led to insights into rare IBMF diseases, including syndromes associated with DHX34, MDM4, RAD51, SRP54, and WIPF1. Our results provide an important population-based framework for the interpretation of LoF variant pathogenicity in rare and emerging IBMF syndromes.
Collapse
|
8
|
Breast-Specific Epigenetic Regulation of DeltaNp73 and Its Role in DNA-Damage-Response of BRCA1-Mutated Human Mammary Epithelial Cells. Cancers (Basel) 2020; 12:cancers12092367. [PMID: 32825620 PMCID: PMC7564633 DOI: 10.3390/cancers12092367] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
The function of BRCA1/2 proteins is essential for maintaining genomic integrity in all cell types. However, why women who carry deleterious germline mutations in BRCA face an extremely high risk of developing breast and ovarian cancers specifically has remained an enigma. We propose that breast-specific epigenetic modifications, which regulate tissue differentiation, could team up with BRCA deficiency and affect tissue susceptibility to cancer. In earlier work, we compared genome-wide methylation profiles of various normal epithelial tissues and identified breast-specific methylated gene promoter regions. Here, we focused on deltaNp73, the truncated isoform of p73, which possesses antiapoptotic and pro-oncogenic functions. We showed that the promoter of deltaNp73 is unmethylated in normal human breast epithelium and methylated in various other normal epithelial tissues and cell types. Accordingly, deltaNp73 was markedly induced by DNA damage in human mammary epithelial cells (HMECs) but not in other epithelial cell types. Moreover, the induction of deltaNp73 protected HMECs from DNA damage-induced cell death, and this effect was more substantial in HMECs from BRCA1 mutation carriers. Notably, when BRCA1 was knocked down in MCF10A, a non-malignant breast epithelial cell line, both deltaNp73 induction and its protective effect from cell death were augmented upon DNA damage. Interestingly, deltaNp73 induction also resulted in inhibition of BRCA1 and BRCA2 expression following DNA damage. In conclusion, breast-specific induction of deltaNp73 promotes survival of BRCA1-deficient mammary epithelial cells upon DNA damage. This might result in the accumulation of genomic alterations and allow the outgrowth of breast cancers. These findings indicate deltaNp73 as a potential modifier of breast cancer susceptibility in BRCA1 mutation carriers and may stimulate novel strategies of prevention and treatment for these high-risk women.
Collapse
|
9
|
Singh AK, Yu X. Tissue-Specific Carcinogens as Soil to Seed BRCA1/2-Mutant Hereditary Cancers. Trends Cancer 2020; 6:559-568. [PMID: 32336659 DOI: 10.1016/j.trecan.2020.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023]
Abstract
Despite their ubiquitous expression, the inheritance of monoallelic germline mutations in breast cancer susceptibility gene type 1 or 2 (BRCA1/2) poses tissue-specific variations in cancer risks and primarily associate with familial breast and ovarian cancers. The molecular basis of this tissue-specific tumor incidence remains unknown and intriguing to cancer researchers. A plethora of recent reports support the idea that several nongenetic factors present in the tissue microenvironment could induce tumors in the mutant BRCA1/2 background. This Opinion article summarizes the recent advances on tissue-specific carcinogens and their complex crosstalk with the compromised DNA repair machinery of BRCA1/2-mutant cells. Finally, we present our perspective on the therapeutic and chemopreventive interpretations of these developments.
Collapse
Affiliation(s)
- Anup Kumar Singh
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Xiaochun Yu
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA.
| |
Collapse
|
10
|
Przybytkowski E, Davis T, Hosny A, Eismann J, Matulonis UA, Wulf GM, Nabavi S. An immune-centric exploration of BRCA1 and BRCA2 germline mutation related breast and ovarian cancers. BMC Cancer 2020; 20:197. [PMID: 32164626 PMCID: PMC7068944 DOI: 10.1186/s12885-020-6605-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/04/2020] [Indexed: 12/28/2022] Open
Abstract
Background BRCA1/2 germline mutation related cancers are candidates for new immune therapeutic interventions. This study was a hypothesis generating exploration of genomic data collected at diagnosis for 19 patients. The prominent tumor mutation burden (TMB) in hereditary breast and ovarian cancers in this cohort was not correlated with high global immune activity in their microenvironments. More information is needed about the relationship between genomic instability, phenotypes and immune microenvironments of these hereditary tumors in order to find appropriate markers of immune activity and the most effective anticancer immune strategies. Methods Mining and statistical analyses of the original DNA and RNA sequencing data and The Cancer Genome Atlas data were performed. To interpret the data, we have used published literature and web available resources such as Gene Ontology, The Cancer immunome Atlas and the Cancer Research Institute iAtlas. Results We found that BRCA1/2 germline related breast and ovarian cancers do not represent a unique phenotypic identity, but they express a range of phenotypes similar to sporadic cancers. All breast and ovarian BRCA1/2 related tumors are characterized by high homologous recombination deficiency (HRD) and low aneuploidy. Interestingly, all sporadic high grade serous ovarian cancers (HGSOC) and most of the subtypes of triple negative breast cancers (TNBC) also express a high degree of HRD. Conclusions TMB is not associated with the magnitude of the immune response in hereditary BRCA1/2 related breast and ovarian cancers or in sporadic TNBC and sporadic HGSOC. Hereditary tumors express phenotypes as heterogenous as sporadic tumors with various degree of “BRCAness” and various characteristics of the immune microenvironments. The subtyping criteria developed for sporadic tumors can be applied for the classification of hereditary tumors and possibly also characterization of their immune microenvironment. A high HRD score may be a good candidate biomarker for response to platinum, and potentially PARP-inhibition. Trial registration Phase I Study of the Oral PI3kinase Inhibitor BKM120 or BYL719 and the Oral PARP Inhibitor Olaparib in Patients With Recurrent TNBC or HGSOC (NCT01623349), first posted on June 20, 2012. The design and the outcome of the clinical trial is not in the scope of this study.
Collapse
Affiliation(s)
- Ewa Przybytkowski
- Department of Computer Science and Engineering, University of Connecticut, Institute of System Genomics, Boston, MA, USA
| | - Thomas Davis
- Department of Computer Science and Engineering, University of Connecticut, Institute of System Genomics, Boston, MA, USA
| | - Abdelrahman Hosny
- Department of Computer Science and Engineering, University of Connecticut, Institute of System Genomics, Boston, MA, USA
| | | | | | - Gerburg M Wulf
- Beth Israel Deaconess Medical Center, Department of Hematology/Oncology, Harvard Medical School, Boston, MA, USA
| | - Sheida Nabavi
- Department of Computer Science and Engineering, University of Connecticut, Institute of System Genomics, Boston, MA, USA.
| |
Collapse
|
11
|
Hilton HN, Patterson McDonald LJ, Santucci N, van der Bent FR, Silvestri A, Graham JD, Clarke CL. BRCA1 Attenuates Progesterone Effects on Proliferation and NFκB Activation in Normal Human Mammary Epithelial Cells. J Mammary Gland Biol Neoplasia 2019; 24:257-270. [PMID: 31104199 DOI: 10.1007/s10911-019-09431-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 04/21/2019] [Indexed: 12/25/2022] Open
Abstract
Germline mutations in the breast cancer susceptibility gene BRCA1, encoding a tumor suppressor protein, greatly enhance the risk of breast and ovarian cancer. This tissue-specificity implicates the role of ovarian hormones. Indeed, BRCA1 has been demonstrated to regulate the signalling axis of the hormone, progesterone, and its receptor, the progesterone receptor (PR), and progesterone action has been implicated in BRCA1-related tumorigenesis. BRCA1 also plays important roles in oxidative stress and activating nuclear factor kappaB (NFκB) signalling pathways. Like wildtype BRCA1 function, PR signalling has also been shown to inhibit NFκB activation. Although PR and BRCA1 networks are known to interact, their interaction at the level of NFκB activation in the human breast is not understood. This study investigates the effect of reduced BRCA1 expression on proliferation and NFκB activation in human breast cells, and the impact of progesterone on these effects. The major findings are that: 1) Reduced BRCA1 levels inhibit cell growth in normal human mammary cells and breast cancer cells; 2) Reduced BRCA1 levels stimulated inflammatory targets and NFκB activity in normal human mammary cells; 3) Wildtype BRCA1 inhibited the pro-proliferative effects of progesterone in normal mammary epithelial cells, and; 4) Progesterone attenuated BRCA1-mediated NFκB activation in normal human mammary cells. These data have important implications for our understanding of progesterone action in BRCA1 mutation carriers, and how inhibition of this action may potentially delay tumorigenesis or impart a more favourable prognosis.
Collapse
Affiliation(s)
- H N Hilton
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney Medical School - Westmead, The University of Sydney, Westmead, NSW, 2145, Australia
| | - L J Patterson McDonald
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney Medical School - Westmead, The University of Sydney, Westmead, NSW, 2145, Australia
| | - N Santucci
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney Medical School - Westmead, The University of Sydney, Westmead, NSW, 2145, Australia
| | - F R van der Bent
- Department of Medicine, Academic Medical Center, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - A Silvestri
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney Medical School - Westmead, The University of Sydney, Westmead, NSW, 2145, Australia
| | - J D Graham
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney Medical School - Westmead, The University of Sydney, Westmead, NSW, 2145, Australia.
| | - C L Clarke
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney Medical School - Westmead, The University of Sydney, Westmead, NSW, 2145, Australia
| |
Collapse
|
12
|
Zong D, Adam S, Wang Y, Sasanuma H, Callén E, Murga M, Day A, Kruhlak MJ, Wong N, Munro M, Chaudhuri AR, Karim B, Xia B, Takeda S, Johnson N, Durocher D, Nussenzweig A. BRCA1 Haploinsufficiency Is Masked by RNF168-Mediated Chromatin Ubiquitylation. Mol Cell 2019; 73:1267-1281.e7. [PMID: 30704900 PMCID: PMC6430682 DOI: 10.1016/j.molcel.2018.12.010] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/22/2018] [Accepted: 12/13/2018] [Indexed: 12/22/2022]
Abstract
BRCA1 functions at two distinct steps during homologous recombination (HR). Initially, it promotes DNA end resection, and subsequently it recruits the PALB2 and BRCA2 mediator complex, which stabilizes RAD51-DNA nucleoprotein filaments. Loss of 53BP1 rescues the HR defect in BRCA1-deficient cells by increasing resection, suggesting that BRCA1's downstream role in RAD51 loading is dispensable when 53BP1 is absent. Here we show that the E3 ubiquitin ligase RNF168, in addition to its canonical role in inhibiting end resection, acts in a redundant manner with BRCA1 to load PALB2 onto damaged DNA. Loss of RNF168 negates the synthetic rescue of BRCA1 deficiency by 53BP1 deletion, and it predisposes BRCA1 heterozygous mice to cancer. BRCA1+/-RNF168-/- cells lack RAD51 foci and are hypersensitive to PARP inhibitor, whereas forced targeting of PALB2 to DNA breaks in mutant cells circumvents BRCA1 haploinsufficiency. Inhibiting the chromatin ubiquitin pathway may, therefore, be a synthetic lethality strategy for BRCA1-deficient cancers.
Collapse
Affiliation(s)
- Dali Zong
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Salomé Adam
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Yifan Wang
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Hiroyuki Sasanuma
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Elsa Callén
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Matilde Murga
- Genomic Instability Group, Spanish National Cancer Research Center, CNIO, Madrid, Spain
| | - Amanda Day
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Michael J. Kruhlak
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Nancy Wong
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Meagan Munro
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Arnab Ray Chaudhuri
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD, USA.,Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Baktiar Karim
- Pathology/Histotechnology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Bing Xia
- Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Shunichi Takeda
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Neil Johnson
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Daniel Durocher
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - André Nussenzweig
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
13
|
Furth PA. Peroxisome proliferator-activated receptor gamma and BRCA1. Endocr Relat Cancer 2019; 26:R73-R79. [PMID: 30444720 PMCID: PMC6494719 DOI: 10.1530/erc-18-0449] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 11/14/2018] [Indexed: 01/02/2023]
Abstract
Peroxisome proliferator-activated receptor gamma agonists have been proposed as breast cancer preventives. Individuals who carry a mutated copy of BRCA1, DNA repair-associated gene, are at increased risk for development of breast cancer. Published data in the field suggest there could be interactions between peroxisome proliferator-activated receptor gamma and BRCA1 that could influence the activity of peroxisome proliferator-activated receptor gamma agonists for prevention. This review explores these possible interactions between peroxisome proliferator-activated receptor gamma, peroxisome proliferator-activated receptor gamma agonists and BRCA1 and discusses feasible experimental directions to provide more definitive information on the potential connections.
Collapse
Affiliation(s)
- Priscilla A Furth
- Departments of Oncology and Medicine, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA
| |
Collapse
|
14
|
Rare variants in Fanconi anemia genes are enriched in acute myeloid leukemia. Blood Cancer J 2018; 8:50. [PMID: 29891941 PMCID: PMC6002376 DOI: 10.1038/s41408-018-0090-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/17/2018] [Accepted: 04/30/2018] [Indexed: 12/12/2022] Open
|
15
|
Böck J, Appenzeller S, Haertle L, Schneider T, Gehrig A, Schröder J, Rost S, Wolf B, Bartram CR, Sutter C, Haaf T. Single CpG hypermethylation, allele methylation errors, and decreased expression of multiple tumor suppressor genes in normal body cells of mutation-negative early-onset and high-risk breast cancer patients. Int J Cancer 2018; 143:1416-1425. [PMID: 29659014 PMCID: PMC6099327 DOI: 10.1002/ijc.31526] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 03/28/2018] [Indexed: 01/01/2023]
Abstract
To evaluate the role of constitutive epigenetic changes in normal body cells of BRCA1/BRCA2-mutation negative patients, we have developed a deep bisulfite sequencing assay targeting the promoter regions of 8 tumor suppressor (TS) genes (BRCA1, BRCA2, RAD51C, ATM, PTEN, TP53, MLH1, RB1) and the estrogene receptor gene (ESR1), which plays a role in tumor progression. We analyzed blood samples of two breast cancer (BC) cohorts with early onset (EO) and high risk (HR) for a heterozygous mutation, respectively, along with age-matched controls. Methylation analysis of up to 50,000 individual DNA molecules per gene and sample allowed quantification of epimutations (alleles with >50% methylated CpGs), which are associated with epigenetic silencing. Compared to ESR1, which is representative for an average promoter, TS genes were characterized by a very low (< 1%) average methylation level and a very low mean epimutation rate (EMR; < 0.0001% to 0.1%). With exception of BRCA1, which showed an increased EMR in BC (0.31% vs. 0.06%), there was no significant difference between patients and controls. One of 36 HR BC patients exhibited a dramatically increased EMR (14.7%) in BRCA1, consistent with a disease-causing epimutation. Approximately one third (15 of 44) EO BC patients exhibited increased rates of single CpG methylation errors in multiple TS genes. Both EO and HR BC patients exhibited global underexpression of blood TS genes. We propose that epigenetic abnormalities in normal body cells are indicative of disturbed mechanisms for maintaining low methylation and appropriate expression levels and may be associated with an increased BC risk.
Collapse
Affiliation(s)
- Julia Böck
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | | | - Larissa Haertle
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Tamara Schneider
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Andrea Gehrig
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Jörg Schröder
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Simone Rost
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Beat Wolf
- University of Applied Sciences Western Switzerland, Fribourg, Switzerland.,Department of Bioinformatics, University of Würzburg, Würzburg, Germany
| | - Claus R Bartram
- Institute of Human Genetics, University Hospital, Heidelberg, Germany
| | - Christian Sutter
- Institute of Human Genetics, University Hospital, Heidelberg, Germany
| | - Thomas Haaf
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| |
Collapse
|
16
|
Tanaka H, Phipps EA, Wei T, Wu X, Goswami C, Liu Y, Sledge GW, Mina L, Herbert BS. Altered expression of telomere-associated genes in leukocytes among BRCA1 and BRCA2 carriers. Mol Carcinog 2018; 57:567-575. [PMID: 29240257 PMCID: PMC5832588 DOI: 10.1002/mc.22773] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/29/2017] [Accepted: 12/07/2017] [Indexed: 12/31/2022]
Abstract
Telomere dysfunction resulting from telomere shortening and deregulation of shelterin components has been linked to the pathogenesis of age-related disorders, including cancer. Recent evidence suggests that BRCA1/2 (BRCA1 and BRCA2) tumor suppressor gene products play an important role in telomere maintenance. Although telomere shortening has been reported in BRCA1/2 carriers, the direct effects of BRCA1/2 haploinsufficiency on telomere maintenance and predisposition to cancer development are not completely understood. In this study, we assessed the telomere-associated and telomere-proximal gene expression profiles in peripheral blood leukocytes from patients with a BRCA1 or BRCA2 mutation, compared to samples from sporadic and familial breast cancer individuals. We found that 25 genes, including TINF2 gene (a negative regulator of telomere length), were significantly differentially expressed in BRCA1 carriers. Leukocyte telomere length analysis revealed that BRCA1/2 carriers had relatively shorter telomeres than healthy controls. Further, affected BRCA1/2 carriers were well differentiated from unaffected BRCA1/2 carriers by the expression of telomere-proximal genes. Our results link BRCA1/2 haploinsufficiency to changes in telomere length, telomere-associated as well as telomere-proximal gene expression. Thus, this work supports the effect of BRCA1/2 haploinsufficiency in the biology underlying telomere dysfunction in cancer development. Future studies evaluating these findings will require a large study population.
Collapse
Affiliation(s)
- Hiromi Tanaka
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN
| | - Elizabeth A. Phipps
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | - Ting Wei
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | - Xi Wu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | - Chirayu Goswami
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN
| | | | - Lida Mina
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Brittney-Shea Herbert
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN
| |
Collapse
|
17
|
Khiabanian H, Hirshfield KM, Goldfinger M, Bird S, Stein M, Aisner J, Toppmeyer D, Wong S, Chan N, Dhar K, Gheeya J, Vig H, Hadigol M, Pavlick D, Ansari S, Ali S, Xia B, Rodriguez-Rodriguez L, Ganesan S. Inference of Germline Mutational Status and Evaluation of Loss of Heterozygosity in High-Depth, Tumor-Only Sequencing Data. JCO Precis Oncol 2018; 2018:PO.17.00148. [PMID: 30246169 PMCID: PMC6148761 DOI: 10.1200/po.17.00148] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Inherited germline defects are implicated in up to 10% of human tumors, with particularly well-known roles in breast and ovarian cancers that harbor BRCA1/2-mutated genes. There is also increasing evidence for the role of germline alterations in other malignancies such as colon and pancreatic cancers. Mutations in familial cancer genes can be detected by high throughput sequencing (HTS), when applied to formalin-fixed paraffin-embedded (FFPE) tumor specimens. However, due to often lack of patient-matched control normal DNA and/or low tumor purity, there is limited ability to determine the genomic status of these alterations (germline versus somatic) and to assess the presence of loss of heterozygosity (LOH). These analyses, especially when applied to genes such as BRCA1/2, can have significant clinical implications for patient care. METHODS LOHGIC (LOH-Germline Inference Calculator) is a statistical model selection method to determine somatic-versus-germline status and predict LOH for mutations identified via clinical grade, high-depth, hybrid-capture tumor-only sequencing. LOHGIC incorporates statistical uncertainties inherent to HTS as well as specimen biases in tumor purity estimates, which we use to assess BRCA1/2 mutations in 1,636 specimens sequenced at Rutgers Cancer Institute of New Jersey. RESULTS Evaluation of LOHGIC with available germline sequencing from BRCA1/2 testing, demonstrates 93% accuracy, 100% precision, and 96% recall. This analysis highlights a differential tumor spectrum associated with BRCA1/2 mutations. CONCLUSION LOHGIC can assess LOH status for both germline and somatic mutations. It also can be applied to any gene with candidate, inherited mutations. This approach demonstrates the clinical utility of targeted sequencing in both identifying patients with potential germline alterations in tumor suppressor genes as well as estimating LOH occurrence in cancer cells, which may confer therapeutic relevance.
Collapse
Affiliation(s)
- Hossein Khiabanian
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Kim M. Hirshfield
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Mendel Goldfinger
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Simon Bird
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Mark Stein
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Joseph Aisner
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Deborah Toppmeyer
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Serena Wong
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Nancy Chan
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Kalyani Dhar
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Jinesh Gheeya
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Hetal Vig
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Mohammad Hadigol
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Dean Pavlick
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Sepand Ansari
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Siraj Ali
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Bing Xia
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Lorna Rodriguez-Rodriguez
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| | - Shridar Ganesan
- Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Simon Bird, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Kalyani Dhar, Jinesh Gheeya, Hetal Vig, Mohammad Hadigol, Sepand Ansari, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Cancer Institute of New Jersey, Rutgers University; Hossein Khiabanian, Kim M. Hirshfield, Mendel Goldfinger, Mark Stein, Joseph Aisner, Deborah Toppmeyer, Serena Wong, Nancy Chan, Bing Xia, Lorna Rodriguez-Rodriguez, and Shridar Ganesan, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ; and Dean Pavlick and Siraj Ali, Foundation Medicine, Cambridge, MA
| |
Collapse
|
18
|
Gómez-Flores-Ramos L, Álvarez-Gómez RM, Villarreal-Garza C, Wegman-Ostrosky T, Mohar A. Breast cancer genetics in young women: What do we know? MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2017; 774:33-45. [PMID: 29173497 DOI: 10.1016/j.mrrev.2017.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 04/21/2017] [Accepted: 08/17/2017] [Indexed: 12/12/2022]
Abstract
Breast cancer (BC) in young women, generally defined in oncology as women who are 40 years of age or younger, represents 2 out of 10 BC cases in developing countries. Several research studies, including genetic cancer panel tests, genome-wide association studies, expression analyses and polymorphisms reports, have found that young women with BC exhibit a higher genetic susceptibility and specific genomic signature compared to postmenopausal women with BC. Thus, international guidelines recommend genetic counseling for this age population. This review presents the current state of the art of genetics and genomics with regards to young women with BC.
Collapse
Affiliation(s)
- Liliana Gómez-Flores-Ramos
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Mario de la Cueva, Coyoacán, Ciudad Universitaria, C.P. 04510, Mexico City, Mexico; Unidad de Investigación en Epidemiología, Subdivisión de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando # 22, Col. Sección XVI, Delegación Tlalpan, C.P. 14080, Mexico City, Mexico
| | - Rosa María Álvarez-Gómez
- Clínica de Cáncer Hereditario, Subdivisión de Investigación Básica, Instituto Nacional de Cancerlogía, Av. San Fernando # 22, Col. Sección XVI, Delegación Tlalpan, C.P. 14080, Mexico City, Mexico
| | - Cynthia Villarreal-Garza
- Clínica de Cáncer Hereditario, Subdivisión de Investigación Básica, Instituto Nacional de Cancerlogía, Av. San Fernando # 22, Col. Sección XVI, Delegación Tlalpan, C.P. 14080, Mexico City, Mexico; Centro de Cáncer de Mama, Tecnológico de Monterrey, Centro Médico Zambrano Hellion, 6° Piso Av. Batallón de San Patricio #112 Col. Real San Agustín, San Pedro Garza García C.P. 66278, Nuevo León, Mexico
| | - Talia Wegman-Ostrosky
- Clínica de Cáncer Hereditario, Subdivisión de Investigación Básica, Instituto Nacional de Cancerlogía, Av. San Fernando # 22, Col. Sección XVI, Delegación Tlalpan, C.P. 14080, Mexico City, Mexico
| | - Alejandro Mohar
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Mario de la Cueva, Coyoacán, Ciudad Universitaria, C.P. 04510, Mexico City, Mexico; Unidad de Investigación en Epidemiología, Subdivisión de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando # 22, Col. Sección XVI, Delegación Tlalpan, C.P. 14080, Mexico City, Mexico.
| |
Collapse
|
19
|
Spira A, Yurgelun MB, Alexandrov L, Rao A, Bejar R, Polyak K, Giannakis M, Shilatifard A, Finn OJ, Dhodapkar M, Kay NE, Braggio E, Vilar E, Mazzilli SA, Rebbeck TR, Garber JE, Velculescu VE, Disis ML, Wallace DC, Lippman SM. Precancer Atlas to Drive Precision Prevention Trials. Cancer Res 2017; 77:1510-1541. [PMID: 28373404 PMCID: PMC6681830 DOI: 10.1158/0008-5472.can-16-2346] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 01/20/2017] [Accepted: 01/20/2017] [Indexed: 02/07/2023]
Abstract
Cancer development is a complex process driven by inherited and acquired molecular and cellular alterations. Prevention is the holy grail of cancer elimination, but making this a reality will take a fundamental rethinking and deep understanding of premalignant biology. In this Perspective, we propose a national concerted effort to create a Precancer Atlas (PCA), integrating multi-omics and immunity - basic tenets of the neoplastic process. The biology of neoplasia caused by germline mutations has led to paradigm-changing precision prevention efforts, including: tumor testing for mismatch repair (MMR) deficiency in Lynch syndrome establishing a new paradigm, combinatorial chemoprevention efficacy in familial adenomatous polyposis (FAP), signal of benefit from imaging-based early detection research in high-germline risk for pancreatic neoplasia, elucidating early ontogeny in BRCA1-mutation carriers leading to an international breast cancer prevention trial, and insights into the intricate germline-somatic-immunity interaction landscape. Emerging genetic and pharmacologic (metformin) disruption of mitochondrial (mt) respiration increased autophagy to prevent cancer in a Li-Fraumeni mouse model (biology reproduced in clinical pilot) and revealed profound influences of subtle changes in mt DNA background variation on obesity, aging, and cancer risk. The elaborate communication between the immune system and neoplasia includes an increasingly complex cellular microenvironment and dynamic interactions between host genetics, environmental factors, and microbes in shaping the immune response. Cancer vaccines are in early murine and clinical precancer studies, building on the recent successes of immunotherapy and HPV vaccine immune prevention. Molecular monitoring in Barrett's esophagus to avoid overdiagnosis/treatment highlights an important PCA theme. Next generation sequencing (NGS) discovered age-related clonal hematopoiesis of indeterminate potential (CHIP). Ultra-deep NGS reports over the past year have redefined the premalignant landscape remarkably identifying tiny clones in the blood of up to 95% of women in their 50s, suggesting that potentially premalignant clones are ubiquitous. Similar data from eyelid skin and peritoneal and uterine lavage fluid provide unprecedented opportunities to dissect the earliest phases of stem/progenitor clonal (and microenvironment) evolution/diversity with new single-cell and liquid biopsy technologies. Cancer mutational signatures reflect exogenous or endogenous processes imprinted over time in precursors. Accelerating the prevention of cancer will require a large-scale, longitudinal effort, leveraging diverse disciplines (from genetics, biochemistry, and immunology to mathematics, computational biology, and engineering), initiatives, technologies, and models in developing an integrated multi-omics and immunity PCA - an immense national resource to interrogate, target, and intercept events that drive oncogenesis. Cancer Res; 77(7); 1510-41. ©2017 AACR.
Collapse
Affiliation(s)
- Avrum Spira
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology and Bioinformatics, Boston University School of Medicine, Boston, Massachusetts
| | - Matthew B Yurgelun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ludmil Alexandrov
- Theoretical Division, Center for Nonlinear Studies, Los Alamos National Laboratory, Los Alamos, New Mexico
| | - Anjana Rao
- Division of Signaling and Gene Expression, La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Rafael Bejar
- Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ali Shilatifard
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Olivera J Finn
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Madhav Dhodapkar
- Department of Hematology and Immunology, Yale Cancer Center, New Haven, Connecticut
| | - Neil E Kay
- Department of Hematology, Mayo Clinic Hospital, Rochester, Minnesota
| | - Esteban Braggio
- Department of Hematology, Mayo Clinic Hospital, Phoenix, Arizona
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarah A Mazzilli
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology and Bioinformatics, Boston University School of Medicine, Boston, Massachusetts
| | - Timothy R Rebbeck
- Division of Hematology and Oncology, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Judy E Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Victor E Velculescu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
- Department of Pathology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Mary L Disis
- Department of Medicine, Center for Translational Medicine in Women's Health, University of Washington, Seattle, Washington
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Scott M Lippman
- Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, California.
| |
Collapse
|