101
|
A P53-Independent DNA Damage Response Suppresses Oncogenic Proliferation and Genome Instability. Cell Rep 2021; 30:1385-1399.e7. [PMID: 32023457 DOI: 10.1016/j.celrep.2020.01.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 10/30/2019] [Accepted: 01/03/2020] [Indexed: 12/20/2022] Open
Abstract
The Mre11-Rad50-Nbs1 complex is a DNA double-strand break sensor that mediates a tumor-suppressive DNA damage response (DDR) in cells undergoing oncogenic stress, yet the mechanisms underlying this effect are poorly understood. Using a genetically inducible primary mammary epithelial cell model, we demonstrate that Mre11 suppresses proliferation and DNA damage induced by diverse oncogenic drivers through a p53-independent mechanism. Breast tumorigenesis models engineered to express a hypomorphic Mre11 allele exhibit increased levels of oncogene-induced DNA damage, R-loop accumulation, and chromosomal instability with a characteristic copy number loss phenotype. Mre11 complex dysfunction is identified in a subset of human triple-negative breast cancers and is associated with increased sensitivity to DNA-damaging therapy and inhibitors of ataxia telangiectasia and Rad3 related (ATR) and poly (ADP-ribose) polymerase (PARP). Thus, deficiencies in the Mre11-dependent DDR drive proliferation and genome instability patterns in p53-deficient breast cancers and represent an opportunity for therapeutic exploitation.
Collapse
|
102
|
Filippova OT, Selenica P, Pareja F, Vahdatinia M, Zhu Y, Pei X, Riaz N, Long Roche K, Chi DS, Abu-Rustum NR, Ellenson LH, Reis-Filho JS, Zamarin D, Weigelt B. Molecular characterization of high-grade serous ovarian cancers occurring in younger and older women. Gynecol Oncol 2021; 161:545-552. [PMID: 33674143 DOI: 10.1016/j.ygyno.2021.02.028] [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: 12/02/2020] [Accepted: 02/19/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVES To determine if the mutational landscapes and genomic features of homologous recombination DNA repair defects (HRD) vary between younger and older patients with high-grade serous ovarian cancer (HGSOC). METHODS Younger and older women were defined as bottom and top age quartiles, respectively. HGSOCs from 15 younger (median 49 years, range 35-53) and 15 older women (median 72 years, range 70-87) were subjected to whole-exome sequencing (WES). For validation, HGSOC WES data were obtained from The Cancer Genome Atlas (TCGA), including 38 younger (median 45 years, range 34-50) and 30 older women (median 74 years, range 68-84). Mutational profiles, BRCA1/2 status, genomic HRD features, and for TCGA cases RNA-sequencing-based HRD transcriptomic signatures were assessed. RESULTS In the institutional cohort, pathogenic germline BRCA1/2 mutations were more frequent in younger (5/15) than older women (0/15, p = 0.042). No somatic BRCA1/2 mutations were identified. HGSOCs from older patients preferentially displayed aging-related mutational signatures and, in contrast to younger patients, harbored CCNE1 amplifications (3/15, 20%). In the TCGA cohort, pathogenic germline BRCA1 (younger 8/38, older 0/30, p = 0.007) but not BRCA2 mutations (young 3/38, older 4/30, p = 0.691) were more frequent in younger patients. Again, no somatic BRCA1/2 mutations were identified. HGSOCs from younger women more frequently displayed genomic features of HRD (all, p < 0.05), a significant HRD gene-signature enrichment, but less frequently CCNE1 amplification (p = 0.05). Immunoreactive CLOVAR subtypes were more common in HGSOCs from younger women, and proliferative subtypes in HGSOCs from older women (p = 0.041). CONCLUSIONS HGSOC patients diagnosed at an older age less frequently harbor pathogenic BRCA1 germline mutations and genomic features of HRD than younger women. Individualized treatment options, particularly pertaining to use of PARP inhibitors, in older women may be warranted.
Collapse
Affiliation(s)
- Olga T Filippova
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fresia Pareja
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mahsa Vahdatinia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yingjie Zhu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xin Pei
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kara Long Roche
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dennis S Chi
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadeem R Abu-Rustum
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lora H Ellenson
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dmitriy Zamarin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
103
|
Ladan MM, van Gent DC, Jager A. Homologous Recombination Deficiency Testing for BRCA-Like Tumors: The Road to Clinical Validation. Cancers (Basel) 2021; 13:1004. [PMID: 33670893 PMCID: PMC7957671 DOI: 10.3390/cancers13051004] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 12/21/2022] Open
Abstract
Germline BRCA mutations result in homologous recombination deficiency (HRD) in hereditary breast and ovarian cancer, as well as several types of sporadic tumors. The HRD phenotype makes these tumors sensitive to DNA double strand break-inducing agents, including poly-(ADP-ribose)-polymerase (PARP) inhibitors. Interestingly, a subgroup of cancers without a BRCA mutation also shows an HRD phenotype. Various methods for selecting patients with HRD tumors beyond BRCA-mutations have been explored. These methods are mainly based on DNA sequencing or functional characteristics of the tumor. We here discuss the various tests and the status of their clinical validation.
Collapse
Affiliation(s)
- Marjolijn M. Ladan
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands;
- Oncode Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Dik C. van Gent
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands;
- Oncode Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands;
| |
Collapse
|
104
|
Dai Y, Wang J, Zhao L, Wang Z, Wang J. Tumor Molecular Features Predict Endometrial Cancer Patients' Survival After Open or Minimally Invasive Surgeries. Front Oncol 2021; 11:634857. [PMID: 33718224 PMCID: PMC7952993 DOI: 10.3389/fonc.2021.634857] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/20/2021] [Indexed: 01/22/2023] Open
Abstract
Background The Cancer Genome Atlas (TCGA) project shed light on the vital role of tumor molecular features in predicting endometrial cancer patients’ prognosis. This study aims to investigate the survival impact of surgical approaches on patients with different genetic alterations. Methods 473 endometrial cancer patients from TCGA database were selected. To analyze the prognostic impact of surgical approach, survival analyses were conducted in patients with different molecular features. Finally, a simplified molecular stratification model was established to select patients suitable for open or minimally invasive surgery (MIS). Results In our cohort, 291 patients received open surgery and 182 received MIS. Molecular features influenced patients’ survival after different surgical approaches. Based on survival analyses, three molecular subtypes were generated, with subtype 1 harboring POLE mutation (POLEmt), microsatellite-instability high (MSI-H), homologous recombination repair (HRR) pathway mutation or MUC16 mutation (MUC16mt); subtype 3 carrying TP53 mutation; and subtype 2 without specific molecular feature. The survival influence of molecular subtypes depended on surgical approaches. In the open surgery cohort, three subtypes showed similar survival outcome, while in the MIS cohort, prognosis varied significantly among three subtypes, with subtype 1 the best and subtype 3 the worst. In stepwise Cox regression, molecular subtype was an independent predictor of recurrence-free survival in patients receiving MIS (p < 0.001). Conclusion The molecular features of endometrial cancer are associated with patients’ prognosis after different surgical approaches. MIS should be recommended in patients with POLEmt, MSI-H, HRR pathway mutation or MUC16mt, while for patients with TP53 mutation, open surgery is better concerning oncological safety.
Collapse
Affiliation(s)
- Yibo Dai
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| | - Jingyuan Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| | - Luyang Zhao
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| | - Zhiqi Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| | - Jianliu Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
105
|
Li LY, Guan YD, Chen XS, Yang JM, Cheng Y. DNA Repair Pathways in Cancer Therapy and Resistance. Front Pharmacol 2021; 11:629266. [PMID: 33628188 PMCID: PMC7898236 DOI: 10.3389/fphar.2020.629266] [Citation(s) in RCA: 210] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 12/31/2020] [Indexed: 12/21/2022] Open
Abstract
DNA repair pathways are triggered to maintain genetic stability and integrity when mammalian cells are exposed to endogenous or exogenous DNA-damaging agents. The deregulation of DNA repair pathways is associated with the initiation and progression of cancer. As the primary anti-cancer therapies, ionizing radiation and chemotherapeutic agents induce cell death by directly or indirectly causing DNA damage, dysregulation of the DNA damage response may contribute to hypersensitivity or resistance of cancer cells to genotoxic agents and targeting DNA repair pathway can increase the tumor sensitivity to cancer therapies. Therefore, targeting DNA repair pathways may be a potential therapeutic approach for cancer treatment. A better understanding of the biology and the regulatory mechanisms of DNA repair pathways has the potential to facilitate the development of inhibitors of nuclear and mitochondria DNA repair pathways for enhancing anticancer effect of DNA damage-based therapy.
Collapse
Affiliation(s)
- Lan-Ya Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Yi-di Guan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xi-Sha Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jin-Ming Yang
- Department of Cancer Biology and Toxicology, Department of Pharmacology, College of Medicine, Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - Yan Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
106
|
Yu KD, Ye FG, He M, Fan L, Ma D, Mo M, Wu J, Liu GY, Di GH, Zeng XH, He PQ, Wu KJ, Hou YF, Wang J, Wang C, Zhuang ZG, Song CG, Lin XY, Toss A, Ricci F, Shen ZZ, Shao ZM. Effect of Adjuvant Paclitaxel and Carboplatin on Survival in Women With Triple-Negative Breast Cancer: A Phase 3 Randomized Clinical Trial. JAMA Oncol 2021; 6:1390-1396. [PMID: 32789480 PMCID: PMC7426881 DOI: 10.1001/jamaoncol.2020.2965] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Question Does a paclitaxel-plus-carboplatin (PCb) as adjuvant treatment in women with operable triple-negative breast cancer offer superior benefit compared with a standard-dose CEF-T regimen (cyclophosphamide, epirubicin, and fluorouracil followed by docetaxel)? Findings In this randomized phase 3 clinical trial conducted at 9 cancer centers and hospitals in China and including 647 patients, after a median follow-up of 62 months, 5-year disease-free survival rate was statistically significantly higher in the PCb group compared with the CEF-T group. Meaning Results of this study suggest that a paclitaxel-plus-carboplatin regimen may be an alternative adjuvant chemotherapy choice for patients with operable triple-negative breast cancer. Importance The value of platinum-based adjuvant chemotherapy in patients with triple-negative breast cancer (TNBC) remains controversial, as does whether BRCA1 and BRCA2 (BRCA1/2) germline variants are associated with platinum treatment sensitivity. Objective To compare 6 cycles of paclitaxel plus carboplatin (PCb) with a standard-dose regimen of 3 cycles of cyclophosphamide, epirubicin, and fluorouracil followed by 3 cycles of docetaxel (CEF-T). Design, Setting, and Participants This phase 3 randomized clinical trial was conducted at 9 cancer centers and hospitals in China. Between July 1, 2011, and April 30, 2016, women aged 18 to 70 years with operable TNBC after definitive surgery (having pathologically confirmed regional node-positive disease or node-negative disease with tumor diameter >10 mm) were screened and enrolled. Exclusion criteria included having metastatic or locally advanced disease, having non-TNBC, or receiving preoperative anticancer therapy. Data were analyzed from December 1, 2019, to January 31, 2020, from the intent-to-treat population as prespecified in the protocol. Interventions Participants were randomized to receive PCb (paclitaxel 80 mg/m2 and carboplatin [area under the curve = 2] on days 1, 8, and 15 every 28 days for 6 cycles) or CEF-T (cyclophosphamide 500 mg/m2, epirubicin 100 mg/m2, and fluorouracil 500 mg/m2 every 3 weeks for 3 cycles followed by docetaxel 100 mg/m2 every 3 weeks for 3 cycles). Main Outcomes and Measures The primary end point was disease-free survival (DFS). Secondary end points included overall survival, distant DFS, relapse-free survival, DFS in patients with germline variants in BRCA1/2 or homologous recombination repair (HRR)–related genes, and toxicity. Results A total of 647 patients (mean [SD] age, 51 [44-57] years) with operable TNBC were randomized to receive CEF-T (n = 322) or PCb (n = 325). At a median follow-up of 62 months, DFS time was longer in those assigned to PCb compared with CEF-T (5-year DFS, 86.5% vs 80.3%, hazard ratio [HR] = 0.65; 95% CI, 0.44-0.96; P = .03). Similar outcomes were observed for distant DFS and relapse-free survival. There was no statistically significant difference in overall survival between the groups (HR = 0.71; 95% CI, 0.42-1.22, P = .22). In the exploratory and hypothesis-generating subgroup analyses of PCb vs CEF-T, the HR for DFS was 0.44 (95% CI, 0.15-1.31; P = .14) in patients with the BRCA1/2 variant and 0.39 (95% CI, 0.15-0.99; P = .04) in those with the HRR variant. Safety data were consistent with the known safety profiles of relevant drugs. Conclusions and Relevance These findings suggest that a paclitaxel-plus-carboplatin regimen is an effective alternative adjuvant chemotherapy choice for patients with operable TNBC. In the era of molecular classification, subsets of TNBC sensitive to PCb should be further investigated. Trial Registration ClinicalTrials.gov Identifier: NCT01216111
Collapse
Affiliation(s)
- Ke-Da Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Fu-Gui Ye
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Min He
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lei Fan
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ding Ma
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Miao Mo
- Department of Cancer Prevention & Clinical Statistics Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jiong Wu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Guang-Yu Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Gen-Hong Di
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiao-Hua Zeng
- Breast Center, Chongqing Cancer Hospital, Chongqing University, Chongqing, China
| | - Ping-Qing He
- Department of Breast Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ke-Jin Wu
- Department of Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yi-Feng Hou
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jie Wang
- Department of Breast Surgery, The International Peace Maternity & Child Health Hospital of China Welfare Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng Wang
- Department of Breast Surgery, Shanghai Ninth People's Hospital Huangpu Branch, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Gang Zhuang
- Department of Breast Surgery, Shanghai First Maternity and Infant Hospital, Shanghai Tongji University, Shanghai, China
| | - Chuan-Gui Song
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiao-Yan Lin
- Department of Breast Surgery, Tongji University School of Medicine Yangpu Hospital, Shanghai, China
| | - Angela Toss
- Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Francesco Ricci
- Department of Drug Development and Innovation, Institute Curie, Paris & Saint-Cloud, France
| | - Zhen-Zhou Shen
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Shanghai, China
| |
Collapse
|
107
|
Yoshida R, Hagio T, Kaneyasu T, Gotoh O, Osako T, Tanaka N, Amino S, Yaguchi N, Nakashima E, Kitagawa D, Ueno T, Ohno S, Nakajima T, Nakamura S, Miki Y, Hirota T, Takahashi S, Matsuura M, Noda T, Mori S. Pathogenicity assessment of variants for breast cancer susceptibility genes based on BRCAness of tumor sample. Cancer Sci 2021; 112:1310-1319. [PMID: 33421217 PMCID: PMC7935793 DOI: 10.1111/cas.14803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/03/2020] [Accepted: 01/05/2021] [Indexed: 12/26/2022] Open
Abstract
Genes involved in the homologous recombination repair pathway—as exemplified by BRCA1, BRCA2, PALB2, ATM, and CHEK2—are frequently associated with hereditary breast and ovarian cancer syndrome. Germline mutations in the loci of these genes with loss of heterozygosity or additional somatic truncation at the WT allele lead to the development of breast cancers with characteristic clinicopathological features and prominent genomic features of homologous recombination deficiency, otherwise referred to as “BRCAness.” Although clinical genetic testing for these and other genes has increased the chances of identifying pathogenic variants, there has also been an increase in the prevalence of variants of uncertain significance, which poses a challenge to patient care because of the difficulties associated with making further clinical decisions. To overcome this challenge, we sought to develop a methodology to reclassify the pathogenicity of these unknown variants using statistical modeling of BRCAness. The model was developed with Lasso logistic regression by comparing 116 genomic attributes derived from 37 BRCA1/2 biallelic mutant and 32 homologous recombination‐quiescent breast cancer exomes. The model showed 95.8% and 86.7% accuracies in the training cohort and The Cancer Genome Atlas validation cohort, respectively. Through application of the model for variant reclassification of homologous recombination‐associated hereditary breast and ovarian cancer causal genes and further assessment with clinicopathological features, we finally identified one likely pathogenic and five likely benign variants. As such, the BRCAness model developed from the tumor exome was robust and provided a reasonable basis for variant reclassification.
Collapse
Affiliation(s)
- Reiko Yoshida
- Department of Oncotherapeutic Medicine, Graduate School of Medicine, Tohoku University, Sendai, Japan.,Division of Cancer Genomics, Cancer Institute, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan.,Department of Clinical Genetic Oncology, Cancer Institute Hospital (CIH), JFCR, Tokyo, Japan
| | - Taichi Hagio
- Division of Cancer Genomics, Cancer Institute, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan.,Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, JFCR, Tokyo, Japan
| | - Tomoko Kaneyasu
- Division of Cancer Genomics, Cancer Institute, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan.,Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, JFCR, Tokyo, Japan
| | - Osamu Gotoh
- Division of Cancer Genomics, Cancer Institute, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan.,Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, JFCR, Tokyo, Japan
| | - Tomo Osako
- Division of Pathology, Cancer Institute, JFCR, Tokyo, Japan
| | - Norio Tanaka
- Division of Cancer Genomics, Cancer Institute, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan.,Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, JFCR, Tokyo, Japan
| | - Sayuri Amino
- Division of Cancer Genomics, Cancer Institute, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan.,Project for Development of Genomics-based Cancer Medicine, Cancer Precision Medicine Center, JFCR, Tokyo, Japan
| | - Noriko Yaguchi
- Division of Cancer Genomics, Cancer Institute, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan.,Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, JFCR, Tokyo, Japan
| | | | - Dai Kitagawa
- Breast Oncology Center, CIH, JFCR, Tokyo, Japan.,Department of Breast Surgical Oncology, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | | | - Shinji Ohno
- Breast Oncology Center, CIH, JFCR, Tokyo, Japan
| | - Takeshi Nakajima
- Department of Clinical Genetic Oncology, Cancer Institute Hospital (CIH), JFCR, Tokyo, Japan
| | - Seigo Nakamura
- Division of Breast Surgical Oncology, Showa University School of Medicine, Tokyo, Japan
| | - Yoshio Miki
- Division of Genetic Diagnosis, Cancer Institute, JFCR, Tokyo, Japan
| | - Toru Hirota
- Department of Cellular and Molecular Imaging of Cancer, Graduate School of Medicine, Tohoku University, Sendai, Japan.,Division of Experimental Pathology, Cancer Institute, JFCR, Tokyo, Japan
| | - Shunji Takahashi
- Department of Oncotherapeutic Medicine, Graduate School of Medicine, Tohoku University, Sendai, Japan.,Department of Medical Oncology, CIH, JFCR, Tokyo, Japan
| | - Masaaki Matsuura
- Division of Cancer Genomics, Cancer Institute, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
| | - Tetsuo Noda
- Division of Cancer Genomics, Cancer Institute, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan.,Project for Development of Genomics-based Cancer Medicine, Cancer Precision Medicine Center, JFCR, Tokyo, Japan.,Cancer, Institute, JFCR, Tokyo, Japan
| | - Seiichi Mori
- Division of Cancer Genomics, Cancer Institute, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan.,Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, JFCR, Tokyo, Japan
| |
Collapse
|
108
|
Kim SJ, Sota Y, Naoi Y, Honma K, Kagara N, Miyake T, Shimoda M, Tanei T, Seno S, Matsuda H, Noguchi S, Shimazu K. Determining homologous recombination deficiency scores with whole exome sequencing and their association with responses to neoadjuvant chemotherapy in breast cancer. Transl Oncol 2021; 14:100986. [PMID: 33340887 PMCID: PMC7750421 DOI: 10.1016/j.tranon.2020.100986] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/28/2020] [Accepted: 12/06/2020] [Indexed: 11/23/2022] Open
Abstract
Recent studies demonstrated that homologous repair deficiency (HRD) score is a useful marker for response to poly (ADP-ribose) polymerase inhibitors or platinum-based chemotherapy. We determined HRD scores and elucidated the clinicopathologic characteristics of HRD-high tumors and their response to non-platinum-based chemotherapy. Primary breast cancer patients (n = 120) were pre-operatively treated with paclitaxel followed by 5-fluorouracil/epirubicin/cyclophosphamide (P-FEC). Germline and somatic homologous recombination related gene mutations (gHRRm and sHRRm, respectively) and HRD scores were analyzed using whole exome sequencing (WES) in tumor tissues obtained before chemotherapy. Of 120 tumors, 30 were determined to be HRD-high tumors, significantly associated with high Ki-67 (P = 0.014), ER negativity (P = 0.007), and PR negativity (P = 0.021). Triple-negative cancers showed significantly higher HRD scores than the luminal, luminal-HER2, and HER2 subtypes (P = 0.023, 0.016, and 0.033, respectively). HRD scores were significantly higher in tumors with gHRRm than in those with sHRRm (P = 0.002) or wild-type HRR genes (P = 1.44e-4), but no significant difference was found in HRD scores between tumors with sHRRm and wild-type HRR genes (P = 0.206). HRD-high tumors had significantly (P = 0.003) higher pCR rates and higher near-pCR rates (P = 0.049) compared with those of the HRD-low tumors in all tumors and the luminal subtype, respectively. HRD-high tumors were associated with aggressive phenotypes and gHRRm, but not sHRRm. Our findings suggested that HRD scores might be useful in predicting response to P-FEC in the luminal subtype.
Collapse
Affiliation(s)
- Seung Jin Kim
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E-10 Yamadaoka, Suita City, Osaka 565-0871, Japan.
| | - Yoshiaki Sota
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E-10 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - Yasuto Naoi
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E-10 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - Keiichiro Honma
- Department of Pathology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka 565-0871, Japan; Department of Diagnostic Pathology and Cytology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka City, Osaka 541-8567, Japan
| | - Naofumi Kagara
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E-10 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - Tomohiro Miyake
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E-10 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - Masafumi Shimoda
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E-10 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - Tomonori Tanei
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E-10 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, 1-5 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - Hideo Matsuda
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, 1-5 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - Shinzaburo Noguchi
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E-10 Yamadaoka, Suita City, Osaka 565-0871, Japan; Hyogo Prefectural Nishinomiya Hospital, 13-9 Rokutanji-cho, Nishinomiya City, Hyogo 662-0918, Japan
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E-10 Yamadaoka, Suita City, Osaka 565-0871, Japan
| |
Collapse
|
109
|
Homologous Recombination Repair Mechanisms in Serous Endometrial Cancer. Cancers (Basel) 2021; 13:cancers13020254. [PMID: 33445465 PMCID: PMC7827019 DOI: 10.3390/cancers13020254] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Serous endometrial cancer is an unusual and aggressive endometrial cancer subtype, conferring the highest mortality of all endometrial cancers. In many ways, it resembles the more common tumor entity high-grade serous ovarian cancer. Thus, there is an urgent need for better treatment options for serous endometrial cancer patients. It is crucial for all dividing cells that the DNA repair is functioning correctly. Our aim was to investigate deficiencies in DNA repair in serous endometrial cancer, in particular the presence of homologous recombination repair deficiency. This kind of DNA repair defect may indicate that a specific targeted therapy, so-called PARP inhibitors, which are already in use for the treatment of ovarian cancer, may be useful also in serous endometrial cancer. This study contributes to the largely unexplored field of DNA repair deficiencies in serous endometrial cancer, and may hence contribute to future improved prognosis for these patients. Abstract Serous endometrial cancer (SEC) resembles high-grade serous ovarian cancer (HGSOC) genetically and clinically, with recurrent copy number alterations, TP53 mutations and a poor prognosis. Thus, SEC patients may benefit from targeted treatments used in HGSOC, e.g., PARP inhibitors. However, the preclinical and clinical knowledge about SEC is scarce, and the exact role of defective DNA repair in this tumor subgroup is largely unknown. We aimed to outline the prevalence of homologous recombination repair deficiency (HRD), copy-number alterations, and somatic mutations in SEC. OncoScan SNP arrays were applied to 19 tumors in a consecutive SEC series to calculate HRD scores and explore global copy-number profiles and genomic aberrations. Copy-number signatures were established and targeted sequencing of 27 HRD-associated genes was performed. All factors were examined in relation to HRD scores to investigate potential drivers of the HRD phenotype. Ten of the 19 SEC tumors (53%) had an HRD score > 42, considered to reflect an HRD phenotype. Higher HRD score was associated with loss of heterozygosity in key HRD genes, and copy-number signatures associated with non-BRCA1/2 dependent HRD in HGSOC. A high number of SECs display an HRD phenotype. It remains to be elucidated whether this also confers PARP inhibitor sensitivity.
Collapse
|
110
|
Vitiello PP, Martini G, Mele L, Giunta EF, De Falco V, Ciardiello D, Belli V, Cardone C, Matrone N, Poliero L, Tirino V, Napolitano S, Della Corte C, Selvaggi F, Papaccio G, Troiani T, Morgillo F, Desiderio V, Ciardiello F, Martinelli E. Vulnerability to low-dose combination of irinotecan and niraparib in ATM-mutated colorectal cancer. J Exp Clin Cancer Res 2021; 40:15. [PMID: 33407715 PMCID: PMC7789007 DOI: 10.1186/s13046-020-01811-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 12/11/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Despite the advancements in new therapies for colorectal cancer (CRC), chemotherapy still constitutes the mainstay of the medical treatment. For this reason, new strategies to increase the efficacy of chemotherapy are desirable. Poly-ADP-Ribose Polymerase inhibitors (PARPi) have shown to increase the activity of DNA damaging chemotherapeutics used in the treatment of CRC, however previous clinical trials failed to validate these results and pointed out dose-limiting toxicities that hamper the use of such combinations in unselected CRC patients. Nevertheless, in these studies little attention was paid to the mutational status of homologous recombination repair (HRR) genes. METHODS We tested the combination of the PARPi niraparib with either 5-fluorouracil, oxaliplatin or irinotecan (SN38) in a panel of 12 molecularly annotated CRC cell lines, encompassing the 4 consensus molecular subtypes (CMSs). Synergism was calculated using the Chou-Talalay method for drug interaction. A correlation between synergism and genetic alterations in genes involved in homologous recombination (HR) repair was performed. We used clonogenic assays, mice xenograft models and patient-derived 3D spheroids to validate the results. The induction of DNA damage was studied by immunofluorescence. RESULTS We showed that human CRC cell lines, as well as patient-derived 3D spheroids, harboring pathogenic ATM mutations are significantly vulnerable to PARPi/chemotherapy combination at low doses, regardless of consensus molecular subtypes (CMS) and microsatellite status. The strongest synergism was shown for the combination of niraparib with irinotecan, and the presence of ATM mutations was associated to a delay in the resolution of double strand breaks (DSBs) through HRR and DNA damage persistence. CONCLUSIONS This work demonstrates that a numerically relevant subset of CRCs carrying heterozygous ATM mutations may benefit from the combination treatment with low doses of niraparib and irinotecan, suggesting a new potential approach in the treatment of ATM-mutated CRC, that deserves to be prospectively validated in clinical trials.
Collapse
Affiliation(s)
- Pietro Paolo Vitiello
- Department of Precision Medicine, Medical Oncology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Giulia Martini
- Department of Precision Medicine, Medical Oncology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Luigi Mele
- Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Emilio Francesco Giunta
- Department of Precision Medicine, Medical Oncology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Vincenzo De Falco
- Department of Precision Medicine, Medical Oncology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Davide Ciardiello
- Department of Precision Medicine, Medical Oncology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Valentina Belli
- Department of Precision Medicine, Medical Oncology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Claudia Cardone
- Department of Precision Medicine, Medical Oncology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Nunzia Matrone
- Department of Precision Medicine, Medical Oncology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Luca Poliero
- Department of Precision Medicine, Medical Oncology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Virginia Tirino
- Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Stefania Napolitano
- Department of Precision Medicine, Medical Oncology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Carminia Della Corte
- Department of Precision Medicine, Medical Oncology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Francesco Selvaggi
- Department of Medical, Surgical, General and oncology surgery, Neurologic, Metabolic and Ageing Sciences, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Gianpaolo Papaccio
- Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Teresa Troiani
- Department of Precision Medicine, Medical Oncology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Floriana Morgillo
- Department of Precision Medicine, Medical Oncology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Vincenzo Desiderio
- Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, Medical Oncology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| | - Erika Martinelli
- Department of Precision Medicine, Medical Oncology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Campania Italy
| |
Collapse
|
111
|
Woo XY, Giordano J, Srivastava A, Zhao ZM, Lloyd MW, de Bruijn R, Suh YS, Patidar R, Chen L, Scherer S, Bailey MH, Yang CH, Cortes-Sanchez E, Xi Y, Wang J, Wickramasinghe J, Kossenkov AV, Rebecca VW, Sun H, Mashl RJ, Davies SR, Jeon R, Frech C, Randjelovic J, Rosains J, Galimi F, Bertotti A, Lafferty A, O’Farrell AC, Modave E, Lambrechts D, ter Brugge P, Serra V, Marangoni E, El Botty R, Kim H, Kim JI, Yang HK, Lee C, Dean DA, Davis-Dusenbery B, Evrard YA, Doroshow JH, Welm AL, Welm BE, Lewis MT, Fang B, Roth JA, Meric-Bernstam F, Herlyn M, Davies MA, Ding L, Li S, Govindan R, Isella C, Moscow JA, Trusolino L, Byrne AT, Jonkers J, Bult CJ, Medico E, Chuang JH. Conservation of copy number profiles during engraftment and passaging of patient-derived cancer xenografts. Nat Genet 2021; 53:86-99. [PMID: 33414553 PMCID: PMC7808565 DOI: 10.1038/s41588-020-00750-6] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 11/18/2020] [Indexed: 02/03/2023]
Abstract
Patient-derived xenografts (PDXs) are resected human tumors engrafted into mice for preclinical studies and therapeutic testing. It has been proposed that the mouse host affects tumor evolution during PDX engraftment and propagation, affecting the accuracy of PDX modeling of human cancer. Here, we exhaustively analyze copy number alterations (CNAs) in 1,451 PDX and matched patient tumor (PT) samples from 509 PDX models. CNA inferences based on DNA sequencing and microarray data displayed substantially higher resolution and dynamic range than gene expression-based inferences, and they also showed strong CNA conservation from PTs through late-passage PDXs. CNA recurrence analysis of 130 colorectal and breast PT/PDX-early/PDX-late trios confirmed high-resolution CNA retention. We observed no significant enrichment of cancer-related genes in PDX-specific CNAs across models. Moreover, CNA differences between patient and PDX tumors were comparable to variations in multiregion samples within patients. Our study demonstrates the lack of systematic copy number evolution driven by the PDX mouse host.
Collapse
Grants
- NC/T001267/1 National Centre for the Replacement, Refinement and Reduction of Animals in Research
- P30 CA016672 NCI NIH HHS
- 29567 Cancer Research UK
- U54 CA233223 NCI NIH HHS
- P30 CA034196 NCI NIH HHS
- P01 CA114046 NCI NIH HHS
- HHSN261201400008C NCI NIH HHS
- P30 CA091842 NCI NIH HHS
- U24 CA224067 NCI NIH HHS
- P50 CA196510 NCI NIH HHS
- U54 CA224070 NCI NIH HHS
- U54 CA224076 NCI NIH HHS
- U54 CA224065 NCI NIH HHS
- U54 CA233306 NCI NIH HHS
- P30 CA010815 NCI NIH HHS
- U24 CA204781 NCI NIH HHS
- U54 CA224083 NCI NIH HHS
- HHSN261201500003C NCI NIH HHS
- HHSN261200800001C NCI NIH HHS
- T32 HG008962 NHGRI NIH HHS
- R50 CA211199 NCI NIH HHS
- P30 CA125123 NCI NIH HHS
- P50 CA070907 NCI NIH HHS
- HHSN261201500003I NCI NIH HHS
- HHSN261200800001E NCI NIH HHS
- P30 CA042014 NCI NIH HHS
- U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- KWF Kankerbestrijding (Dutch Cancer Society)
- Oncode Institute
- Fondazione AIRC under 5 per Mille 2018 - ID. 21091 EU H2020 Research and Innovation Programme, grant agreement no. 731105 European Research Council Consolidator Grant 724748
- EU H2020 Research and Innovation Programme, grant Agreement No. 754923
- EU H2020 Research and Innovation Programme, grant agreement no. 731105 ISCIII - Miguel Servet program CP14/00228 GHD-Pink/FERO Foundation grant
- Fondazione Piemontese per la Ricerca sul Cancro-ONLUS 5 per mille Ministero della Salute 2015
- Korean Health Industry Development Institute HI13C2148
- Korean Health Industry Development Institute HI13C2148 The First Affiliated Hospital of Xi’an Jiaotong University Ewha Womans University Research Grant
- CPRIT RP170691
- SCU | Ignatian Center for Jesuit Education, Santa Clara University
- Breast Cancer Research Foundation (BCRF)
- Fashion Footwear Charitable Foundation of New York The Foundation for Barnes-Jewish Hospital’s Cancer Frontier Fund
- My First AIRC Grant 19047
- Fondazione AIRC under 5 per Mille 2018 - ID. 21091 AIRC Investigator Grants 18532 and 20697 AIRC/CRUK/FC AECC Accelerator Award 22795 Fondazione Piemontese per la Ricerca sul Cancro-ONLUS 5 per mille Ministero della Salute 2015, 2014, 2016 EU H2020 Research and Innovation Programme, grant Agreement No. 754923 EU H2020 Research and Innovation Programme, grant agreement no. 731105
- Science Foundation Ireland (SFI)
- EU H2020 Research and Innovation Programme, grant agreement no. 731105 EU H2020 Research and Innovation Programme, grant Agreement No. 754923 Irish Health Research Board grant ILP-POR-2019-066
- Nederlandse Organisatie voor Wetenschappelijk Onderzoek (Netherlands Organisation for Scientific Research)
- EU H2020 Research and Innovation Programme, grant agreement no. 731105 European Research Council (ERC) Synergy project CombatCancer Oncode Institute
Collapse
Affiliation(s)
- Xing Yi Woo
- grid.249880.f0000 0004 0374 0039The Jackson Laboratory for Genomic Medicine, Farmington, CT USA
| | - Jessica Giordano
- grid.7605.40000 0001 2336 6580Department of Oncology, University of Turin, Turin, Italy ,grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Anuj Srivastava
- grid.249880.f0000 0004 0374 0039The Jackson Laboratory for Genomic Medicine, Farmington, CT USA
| | - Zi-Ming Zhao
- grid.249880.f0000 0004 0374 0039The Jackson Laboratory for Genomic Medicine, Farmington, CT USA
| | - Michael W. Lloyd
- grid.249880.f0000 0004 0374 0039The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME USA
| | - Roebi de Bruijn
- grid.430814.aNetherlands Cancer Institute, Amsterdam, the Netherlands
| | - Yun-Suhk Suh
- grid.31501.360000 0004 0470 5905College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Rajesh Patidar
- grid.418021.e0000 0004 0535 8394Frederick National Laboratory for Cancer Research, Frederick, MD USA
| | - Li Chen
- grid.418021.e0000 0004 0535 8394Frederick National Laboratory for Cancer Research, Frederick, MD USA
| | - Sandra Scherer
- grid.223827.e0000 0001 2193 0096Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT USA
| | - Matthew H. Bailey
- grid.223827.e0000 0001 2193 0096Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT USA ,grid.223827.e0000 0001 2193 0096Department of Human Genetics, University of Utah, Salt Lake City, UT USA
| | - Chieh-Hsiang Yang
- grid.223827.e0000 0001 2193 0096Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT USA
| | - Emilio Cortes-Sanchez
- grid.223827.e0000 0001 2193 0096Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT USA
| | - Yuanxin Xi
- grid.240145.60000 0001 2291 4776Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Jing Wang
- grid.240145.60000 0001 2291 4776Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | | | | | - Vito W. Rebecca
- grid.251075.40000 0001 1956 6678The Wistar Institute, Philadelphia, PA USA
| | - Hua Sun
- grid.4367.60000 0001 2355 7002Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO USA
| | - R. Jay Mashl
- grid.4367.60000 0001 2355 7002Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO USA
| | - Sherri R. Davies
- grid.4367.60000 0001 2355 7002Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO USA
| | - Ryan Jeon
- grid.492568.4Seven Bridges Genomics, Charlestown, MA USA
| | | | | | | | - Francesco Galimi
- grid.7605.40000 0001 2336 6580Department of Oncology, University of Turin, Turin, Italy ,grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Andrea Bertotti
- grid.7605.40000 0001 2336 6580Department of Oncology, University of Turin, Turin, Italy ,grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Adam Lafferty
- grid.4912.e0000 0004 0488 7120Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Alice C. O’Farrell
- grid.4912.e0000 0004 0488 7120Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Elodie Modave
- grid.5596.f0000 0001 0668 7884Center for Cancer Biology, VIB, Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Laboratory of Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Diether Lambrechts
- grid.5596.f0000 0001 0668 7884Center for Cancer Biology, VIB, Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Laboratory of Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Petra ter Brugge
- grid.430814.aNetherlands Cancer Institute, Amsterdam, the Netherlands
| | - Violeta Serra
- grid.411083.f0000 0001 0675 8654Vall d´Hebron Institute of Oncology, Barcelona, Spain
| | - Elisabetta Marangoni
- grid.418596.70000 0004 0639 6384Department of Translational Research, Institut Curie, PSL Research University, Paris, France
| | - Rania El Botty
- grid.418596.70000 0004 0639 6384Department of Translational Research, Institut Curie, PSL Research University, Paris, France
| | - Hyunsoo Kim
- grid.249880.f0000 0004 0374 0039The Jackson Laboratory for Genomic Medicine, Farmington, CT USA
| | - Jong-Il Kim
- grid.31501.360000 0004 0470 5905College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Han-Kwang Yang
- grid.31501.360000 0004 0470 5905College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Charles Lee
- grid.249880.f0000 0004 0374 0039The Jackson Laboratory for Genomic Medicine, Farmington, CT USA ,grid.452438.cPrecision Medicine Center, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China ,grid.255649.90000 0001 2171 7754Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Dennis A. Dean
- grid.492568.4Seven Bridges Genomics, Charlestown, MA USA
| | | | - Yvonne A. Evrard
- grid.418021.e0000 0004 0535 8394Frederick National Laboratory for Cancer Research, Frederick, MD USA
| | - James H. Doroshow
- grid.48336.3a0000 0004 1936 8075Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD USA
| | - Alana L. Welm
- grid.223827.e0000 0001 2193 0096Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT USA
| | - Bryan E. Welm
- grid.223827.e0000 0001 2193 0096Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT USA ,grid.223827.e0000 0001 2193 0096Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT USA
| | - Michael T. Lewis
- grid.39382.330000 0001 2160 926XLester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX USA
| | - Bingliang Fang
- grid.240145.60000 0001 2291 4776Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Jack A. Roth
- grid.240145.60000 0001 2291 4776Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Funda Meric-Bernstam
- grid.240145.60000 0001 2291 4776Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Meenhard Herlyn
- grid.251075.40000 0001 1956 6678The Wistar Institute, Philadelphia, PA USA
| | - Michael A. Davies
- grid.240145.60000 0001 2291 4776Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Li Ding
- grid.4367.60000 0001 2355 7002Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO USA
| | - Shunqiang Li
- grid.4367.60000 0001 2355 7002Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO USA
| | - Ramaswamy Govindan
- grid.4367.60000 0001 2355 7002Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO USA
| | - Claudio Isella
- grid.7605.40000 0001 2336 6580Department of Oncology, University of Turin, Turin, Italy ,grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Jeffrey A. Moscow
- grid.48336.3a0000 0004 1936 8075Investigational Drug Branch, National Cancer Institute, Bethesda, MD USA
| | - Livio Trusolino
- grid.7605.40000 0001 2336 6580Department of Oncology, University of Turin, Turin, Italy ,grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Annette T. Byrne
- grid.4912.e0000 0004 0488 7120Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jos Jonkers
- grid.430814.aNetherlands Cancer Institute, Amsterdam, the Netherlands
| | - Carol J. Bult
- grid.249880.f0000 0004 0374 0039The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME USA
| | - Enzo Medico
- grid.7605.40000 0001 2336 6580Department of Oncology, University of Turin, Turin, Italy ,grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Jeffrey H. Chuang
- grid.249880.f0000 0004 0374 0039The Jackson Laboratory for Genomic Medicine, Farmington, CT USA
| | | | | |
Collapse
|
112
|
Grandal B, Evrevin C, Laas E, Jardin I, Rozette S, Laot L, Dumas E, Coussy F, Pierga JY, Brain E, Saule C, Stoppa-Lyonnet D, Frank S, Sénéchal C, Lae M, De Croze D, Bataillon G, Guerin J, Reyal F, Hamy AS. Impact of BRCA Mutation Status on Tumor Infiltrating Lymphocytes (TILs), Response to Treatment, and Prognosis in Breast Cancer Patients Treated with Neoadjuvant Chemotherapy. Cancers (Basel) 2020; 12:cancers12123681. [PMID: 33302444 PMCID: PMC7764707 DOI: 10.3390/cancers12123681] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Five to 10% of breast cancers (BCs) occur in a genetic predisposition context (mainly BRCA pathogenic variant). Nevertheless, little is known about immune tumor infiltration, response to neoadjuvant chemotherapy (NAC), pathologic complete response (pCR) and adverse events according to BRCA status. MATERIAL AND METHODS Out of 1199 invasive BC patients treated with NAC between 2002 and 2012, we identified 267 patients tested for a germline BRCA pathogenic variant. We evaluated pre-NAC and post-NAC immune infiltration (TILs). Response to chemotherapy was assessed by pCR rates. Association of clinical and pathological factors with TILs, pCR and survival was assessed by univariate and multivariate analyses. RESULTS Among 1199 BC patients: 46 were BRCA-deficient and 221 BRCA-proficient or wild type (WT). At NAC completion, pCR was observed in 84/266 (31%) patients and pCR rates were significantly higher in BRCA-deficient BC (p = 0.001), and this association remained statistically significant only in the luminal BC subtype (p = 0.006). The interaction test between BC subtype and BRCA status was nearly significant (Pinteraction = 0.056). Pre and post-NAC TILs were not significantly different between BRCA-deficient and BRCA-proficient carriers; however, in the luminal BC group, post-NAC TILs were significantly higher in BRCA-deficient BC. Survival analysis were not different between BRCA-carriers and non-carriers. CONCLUSIONS BRCA mutation status is associated with higher pCR rates and post-NAC TILs in patients with luminal BC. BRCA-carriers with luminal BCs may represent a subset of patients deriving higher benefit from NAC. Second line therapies, including immunotherapy after NAC, could be of interest in non-responders to NAC.
Collapse
Affiliation(s)
- Beatriz Grandal
- Department of Surgery, Institut Curie, University Paris, 75005 Paris, France; (B.G.); (C.E.); (E.L.); (I.J.); (S.R.); (L.L.)
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Institut Curie, 26 rue d’Ulm, 75005 Paris, France; (E.D.); (A.-S.H.)
| | - Clémence Evrevin
- Department of Surgery, Institut Curie, University Paris, 75005 Paris, France; (B.G.); (C.E.); (E.L.); (I.J.); (S.R.); (L.L.)
| | - Enora Laas
- Department of Surgery, Institut Curie, University Paris, 75005 Paris, France; (B.G.); (C.E.); (E.L.); (I.J.); (S.R.); (L.L.)
| | - Isabelle Jardin
- Department of Surgery, Institut Curie, University Paris, 75005 Paris, France; (B.G.); (C.E.); (E.L.); (I.J.); (S.R.); (L.L.)
| | - Sonia Rozette
- Department of Surgery, Institut Curie, University Paris, 75005 Paris, France; (B.G.); (C.E.); (E.L.); (I.J.); (S.R.); (L.L.)
| | - Lucie Laot
- Department of Surgery, Institut Curie, University Paris, 75005 Paris, France; (B.G.); (C.E.); (E.L.); (I.J.); (S.R.); (L.L.)
| | - Elise Dumas
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Institut Curie, 26 rue d’Ulm, 75005 Paris, France; (E.D.); (A.-S.H.)
| | - Florence Coussy
- Department of Oncology, Institut Curie, 26 rue d’Ulm, 75005 Paris, France; (F.C.); (J.-Y.P.)
| | - Jean-Yves Pierga
- Department of Oncology, Institut Curie, 26 rue d’Ulm, 75005 Paris, France; (F.C.); (J.-Y.P.)
| | - Etienne Brain
- Department of Oncology, Centre René Huguenin, Institut Curie, 35 rue Dailly, 92210 St Cloud, France;
| | - Claire Saule
- Department of Genetics, Institut Curie, 26 rue d’Ulm, 75005 Paris, France; (C.S.); (D.S.-L.); (S.F.)
| | - Dominique Stoppa-Lyonnet
- Department of Genetics, Institut Curie, 26 rue d’Ulm, 75005 Paris, France; (C.S.); (D.S.-L.); (S.F.)
| | - Sophie Frank
- Department of Genetics, Institut Curie, 26 rue d’Ulm, 75005 Paris, France; (C.S.); (D.S.-L.); (S.F.)
| | - Claire Sénéchal
- Department of Genetics, Institut Bergonié, 229 Cours de l’Argonne, 33000 Bordeaux, France;
| | - Marick Lae
- Department of Pathology, Centre René Huguenin, Institut Curie, 35 rue Dailly, 92210 St Cloud, France; (M.L.); (D.D.C.)
- Department of Pathology, Centre Henri Becquerel, INSERM U1245, UNIROUEN, University of Normandie, 76038 Rouen, France
| | - Diane De Croze
- Department of Pathology, Centre René Huguenin, Institut Curie, 35 rue Dailly, 92210 St Cloud, France; (M.L.); (D.D.C.)
| | | | - Julien Guerin
- Data Office, Institut Curie, 25 rue d’Ulm, 75005 Paris, France;
| | - Fabien Reyal
- Department of Surgery, Institut Curie, University Paris, 75005 Paris, France; (B.G.); (C.E.); (E.L.); (I.J.); (S.R.); (L.L.)
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Institut Curie, 26 rue d’Ulm, 75005 Paris, France; (E.D.); (A.-S.H.)
- Correspondence: ; Tel.: +33-144324660; Fax: +33-153104037
| | - Anne-Sophie Hamy
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Institut Curie, 26 rue d’Ulm, 75005 Paris, France; (E.D.); (A.-S.H.)
| |
Collapse
|
113
|
Jiang M, Jia K, Wang L, Li W, Chen B, Liu Y, Wang H, Zhao S, He Y, Zhou C. Alterations of DNA damage repair in cancer: from mechanisms to applications. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1685. [PMID: 33490197 PMCID: PMC7812211 DOI: 10.21037/atm-20-2920] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
DNA damage repair (DDR) pathways are essential to ensure the accurate transmission of genetic material. However, different endogenous and exogenous factors challenge genomic integrity. Mechanisms involved in the alterations of DDR pathways mainly include genetic inactivation and epigenetic mechanisms. The development and progression of carcinomas are closely associated with DDR pathway aberrations, including the epigenetic silencing of gene O6-alkylguanine-DNA methyltransferase (MGMT); deficiencies of mismatch repair (MMR) genes, including MutL homolog 1 (MLH1), MutS protein homologue (MSH)-2 (MSH2), MSH6, and PMS1 homolog 2; the mismatch repair system component (PMS2); and mutations of homologous recombination repair (HRR) genes, such as the breast cancer susceptibility gene 1/2 (BRCA1/2). Understanding the underlying mechanisms and the correlations between alterations to DDR pathways and cancer could improve the efficacy of antitumor therapies. Emerging evidence suggests that survival is higher in patients with DDR-deficient tumors than in those with DDR-proficient tumors. Thus, DDR alterations play a predictive and prognostic role in anticancer therapies. Theoretical studies on the co-administration of DDR inhibitors and other anticancer therapies, including chemotherapy, radiotherapy, immunotherapy, endocrine therapy, and epigenetic drugs, hold promise for cancer treatments. In this review, we focus on the basic mechanisms, characteristics, current applications, and combination strategies of DDR pathways in the anticancer field.
Collapse
Affiliation(s)
- Minlin Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Tongji University, Shanghai, China
| | - Keyi Jia
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Tongji University, Shanghai, China
| | - Lei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Wei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Bin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yu Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Tongji University, Shanghai, China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Tongji University, Shanghai, China
| | - Sha Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
114
|
Principe DR, Narbutis M, Koch R, Rana A. Frequency and prognostic value of mutations associated with the homologous recombination DNA repair pathway in a large pan cancer cohort. Sci Rep 2020; 10:20223. [PMID: 33214570 PMCID: PMC7677533 DOI: 10.1038/s41598-020-76975-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 10/27/2020] [Indexed: 12/18/2022] Open
Abstract
PARP inhibitors have shown remarkable efficacy in the clinical management of several BRCA-mutated tumors. This approach is based on the long-standing hypothesis that PARP inhibition will impair the repair of single stranded breaks, causing synthetic lethality in tumors with loss of high-fidelity double-strand break homologous recombination. While this is now well accepted and has been the basis of several successful clinical trials, emerging evidence strongly suggests that mutation to several additional genes involved in homologous recombination may also have predictive value for PARP inhibitors. While this notion is supported by early clinical evidence, the mutation frequencies of these and other functionally related genes are largely unknown, particularly in cancers not classically associated with homologous recombination deficiency. We therefore evaluated the mutation status of 22 genes associated with the homologous recombination DNA repair pathway or PARP inhibitor sensitivity, first in a pan-cancer cohort of 55,586 patients, followed by a more focused analysis in The Cancer Genome Atlas cohort of 12,153 patients. In both groups we observed high rates of mutations in a variety of HR-associated genes largely unexplored in the setting of PARP inhibition, many of which were associated also with poor clinical outcomes. We then extended our study to determine which mutations have a known oncogenic role, as well as similar to known oncogenic mutations that may have a similar phenotype. Finally, we explored the individual cancer histologies in which these genomic alterations are most frequent. We concluded that the rates of deleterious mutations affecting genes associated with the homologous recombination pathway may be underrepresented in a wide range of human cancers, and several of these genes warrant further and more focused investigation, particularly in the setting of PARP inhibition and HR deficiency.
Collapse
Affiliation(s)
- Daniel R Principe
- Division of Surgical Oncology, Department of Surgery, College of Medicine, The University of Illinois at Chicago, 840 S. Wood Street, Suite 601 Clinical Sciences Building, Chicago, IL, 60612, USA
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL, USA
| | - Matthew Narbutis
- Division of Surgical Oncology, Department of Surgery, College of Medicine, The University of Illinois at Chicago, 840 S. Wood Street, Suite 601 Clinical Sciences Building, Chicago, IL, 60612, USA
| | - Regina Koch
- University of Illinois College of Medicine, Chicago, IL, USA
| | - Ajay Rana
- Division of Surgical Oncology, Department of Surgery, College of Medicine, The University of Illinois at Chicago, 840 S. Wood Street, Suite 601 Clinical Sciences Building, Chicago, IL, 60612, USA.
- Jesse Brown VA Medical Center, Chicago, IL, USA.
| |
Collapse
|
115
|
Mutations in BRCA1 and BRCA2 differentially affect the tumor microenvironment and response to checkpoint blockade immunotherapy. ACTA ACUST UNITED AC 2020; 1:1188-1203. [PMID: 33834176 DOI: 10.1038/s43018-020-00139-8] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Immune checkpoint blockade (ICB) has improved outcomes for patients with advanced cancer, but the determinants of response remain poorly understood. Here we report differential effects of mutations in the homologous recombination genes BRCA1 and BRCA2 on response to ICB in mouse and human tumors, and further show that truncating mutations in BRCA2 are associated with superior response compared to those in BRCA1. Mutations in BRCA1 and BRCA2 result in distinct mutational landscapes and differentially modulate the tumor-immune microenvironment, with gene expression programs related to both adaptive and innate immunity enriched in BRCA2-deficient tumors. Single-cell RNA sequencing further revealed distinct T cell, natural killer, macrophage, and dendritic cell populations enriched in BRCA2-deficient tumors. Taken together, our findings reveal the divergent effects of BRCA1 and BRCA2-deficiency on ICB outcome, and have significant implications for elucidating the genetic and microenvironmental determinants of response to immunotherapy.
Collapse
|
116
|
Marra A, Trapani D, Viale G, Criscitiello C, Curigliano G. Practical classification of triple-negative breast cancer: intratumoral heterogeneity, mechanisms of drug resistance, and novel therapies. NPJ Breast Cancer 2020; 6:54. [PMID: 33088912 PMCID: PMC7568552 DOI: 10.1038/s41523-020-00197-2] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is not a unique disease, encompassing multiple entities with marked histopathological, transcriptomic and genomic heterogeneity. Despite several efforts, transcriptomic and genomic classifications have remained merely theoretic and most of the patients are being treated with chemotherapy. Driver alterations in potentially targetable genes, including PIK3CA and AKT, have been identified across TNBC subtypes, prompting the implementation of biomarker-driven therapeutic approaches. However, biomarker-based treatments as well as immune checkpoint inhibitor-based immunotherapy have provided contrasting and limited results so far. Accordingly, a better characterization of the genomic and immune contexture underpinning TNBC, as well as the translation of the lessons learnt in the metastatic disease to the early setting would improve patients' outcomes. The application of multi-omics technologies, biocomputational algorithms, assays for minimal residual disease monitoring and novel clinical trial designs are strongly warranted to pave the way toward personalized anticancer treatment for patients with TNBC.
Collapse
Affiliation(s)
- Antonio Marra
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Dario Trapani
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
| | - Giulia Viale
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
| | - Carmen Criscitiello
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| |
Collapse
|
117
|
The genomic landscape of metastatic histologic special types of invasive breast cancer. NPJ Breast Cancer 2020; 6:53. [PMID: 33083532 PMCID: PMC7560857 DOI: 10.1038/s41523-020-00195-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Histologic special types of breast cancer (BC) account for ~20% of BCs. Large sequencing studies of metastatic BC have focused on invasive ductal carcinomas of no special type (IDC-NSTs). We sought to define the repertoire of somatic genetic alterations of metastatic histologic special types of BC. We reanalyzed targeted capture sequencing data of 309 special types of BC, including metastatic and primary invasive lobular carcinomas (ILCs; n = 132 and n = 127, respectively), mixed mucinous (n = 5 metastatic and n = 14 primary), micropapillary (n = 12 metastatic and n = 8 primary), and metaplastic BCs (n = 6 metastatic and n = 5 primary), and compared metastatic histologic special types of BC to metastatic IDC-NSTs matched according to clinicopathologic characteristics and to primary special type BCs. The genomic profiles of metastatic and primary special types of BC were similar. Important differences, however, were noted: metastatic ILCs harbored a higher frequency of genetic alterations in TP53, ESR1, FAT1, RFWD2, and NF1 than primary ILCs, and in CDH1, PIK3CA, ERBB2, TBX3, NCOR1, and RFWD2 than metastatic IDC-NSTs. Metastatic ILCs displayed a higher mutational burden, and more frequently dominant APOBEC mutational signatures than primary ILCs and matched metastatic IDC-NSTs. ESR1 and NCOR mutations were frequently detected in metastatic mixed mucinous BCs, whereas PIK3CA and TP53 were the most frequently altered genes in metastatic micropapillary and metaplastic BCs, respectively. Taken together, primary and metastatic BCs histologic special types have remarkably similar repertoires of somatic genetic alterations. Metastatic ILCs more frequently harbor APOBEC mutational signatures than primary ILCs and metastatic IDC-NSTs.
Collapse
|
118
|
Bodily WR, Shirts BH, Walsh T, Gulsuner S, King MC, Parker A, Roosan M, Piccolo SR. Effects of germline and somatic events in candidate BRCA-like genes on breast-tumor signatures. PLoS One 2020; 15:e0239197. [PMID: 32997669 PMCID: PMC7526916 DOI: 10.1371/journal.pone.0239197] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/02/2020] [Indexed: 11/19/2022] Open
Abstract
Mutations in BRCA1 and BRCA2 cause deficiencies in homologous recombination repair (HR), resulting in repair of DNA double-strand breaks by the alternative non-homologous end-joining pathway, which is more error prone. HR deficiency of breast tumors is important because it is associated with better responses to platinum salt therapies and PARP inhibitors. Among other consequences of HR deficiency are characteristic somatic-mutation signatures and gene-expression patterns. The term "BRCA-like" (or "BRCAness") describes tumors that harbor an HR defect but have no detectable germline mutation in BRCA1 or BRCA2. A better understanding of the genes and molecular events associated with tumors being BRCA-like could provide mechanistic insights and guide development of targeted treatments. Using data from The Cancer Genome Atlas (TCGA) for 1101 breast-cancer patients, we identified individuals with a germline mutation, somatic mutation, homozygous deletion, and/or hypermethylation event in BRCA1, BRCA2, and 59 other cancer-predisposition genes. Based on the assumption that BRCA-like events would have similar downstream effects on tumor biology as BRCA1/BRCA2 germline mutations, we quantified these effects based on somatic-mutation signatures and gene-expression profiles. We reduced the dimensionality of the somatic-mutation signatures and expression data and used a statistical resampling approach to quantify similarities among patients who had a BRCA1/BRCA2 germline mutation, another type of aberration in BRCA1 or BRCA2, or any type of aberration in one of the other genes. Somatic-mutation signatures of tumors having a non-germline aberration in BRCA1/BRCA2 (n = 80) were generally similar to each other and to tumors from BRCA1/BRCA2 germline carriers (n = 44). Additionally, somatic-mutation signatures of tumors with germline or somatic events in ATR (n = 16) and BARD1 (n = 8) showed high similarity to tumors from BRCA1/BRCA2 carriers. Other genes (CDKN2A, CTNNA1, PALB2, PALLD, PRSS1, SDHC) also showed high similarity but only for a small number of events or for a single event type. Tumors with germline mutations or hypermethylation of BRCA1 had relatively similar gene-expression profiles and overlapped considerably with the Basal-like subtype; but the transcriptional effects of the other events lacked consistency. Our findings confirm previously known relationships between molecular signatures and germline or somatic events in BRCA1/BRCA2. Our methodology represents an objective way to identify genes that have similar downstream effects on molecular signatures when mutated, deleted, or hypermethylated.
Collapse
Affiliation(s)
- Weston R. Bodily
- Department of Biology, Brigham Young University, Provo, UT, United States of America
| | - Brian H. Shirts
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Tom Walsh
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Suleyman Gulsuner
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Mary-Claire King
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Alyssa Parker
- Department of Biology, Brigham Young University, Provo, UT, United States of America
| | - Moom Roosan
- Pharmacy Practice Department, Chapman University School of Pharmacy, Irvine, CA, United States of America
| | - Stephen R. Piccolo
- Department of Biology, Brigham Young University, Provo, UT, United States of America
| |
Collapse
|
119
|
Boonen RACM, Vreeswijk MPG, van Attikum H. Functional Characterization of PALB2 Variants of Uncertain Significance: Toward Cancer Risk and Therapy Response Prediction. Front Mol Biosci 2020; 7:169. [PMID: 33195396 PMCID: PMC7525363 DOI: 10.3389/fmolb.2020.00169] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
In recent years it has become clear that pathogenic variants in PALB2 are associated with a high risk for breast, ovarian and pancreatic cancer. However, the clinical relevance of variants of uncertain significance (VUS) in PALB2, which are increasingly identified through clinical genetic testing, is unclear. Here we review recent advances in the functional characterization of VUS in PALB2. A combination of assays has been used to assess the impact of PALB2 VUS on its function in DNA repair by homologous recombination, cell cycle regulation and the control of cellular levels of reactive oxygen species (ROS). We discuss the outcome of this comprehensive analysis of PALB2 VUS, which showed that VUS in PALB2’s Coiled-Coil (CC) domain can impair the interaction with BRCA1, whereas VUS in its WD40 domain affect PALB2 protein stability. Accordingly, the CC and WD40 domains of PALB2 represent hotspots for variants that impair PALB2 protein function. We also provide a future perspective on the high-throughput analysis of VUS in PALB2, as well as the functional characterization of variants that affect PALB2 RNA splicing. Finally, we discuss how results from these functional assays can be valuable for predicting cancer risk and responsiveness to cancer therapy, such as treatment with PARP inhibitor- or platinum-based chemotherapy.
Collapse
Affiliation(s)
- Rick A C M Boonen
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Maaike P G Vreeswijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Haico van Attikum
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
120
|
Sharma A, Almasan A. USP14 Regulates DNA Damage Response and Is a Target for Radiosensitization in Non-Small Cell Lung Cancer. Int J Mol Sci 2020; 21:E6383. [PMID: 32887472 PMCID: PMC7503721 DOI: 10.3390/ijms21176383] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 08/29/2020] [Accepted: 09/01/2020] [Indexed: 01/09/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) represents ~85% of the lung cancer cases. Despite recent advances in NSCLC treatment, the five-year survival rate is still around 23%. Radiotherapy is indicated in the treatment of both early and advanced stage NSCLC; however, treatment response in patients is heterogeneous. Thus, identification of new and more effective treatment combinations is warranted. We have identified Ubiquitin-specific protease 14 (USP14) s a regulator of major double-strand break (DSB) repair pathways in response to ionizing radiation (IR) by its impact on both non-homologous end joining (NHEJ) and homologous recombination (HR) in NSCLC. USP14 is a proteasomal deubiquitinase. IR treatment increases levels and DSB recruitment of USP14 in NSCLC cell lines. Genetic knockdown, using shUSP14 expression or pharmacological inhibition of USP14, using IU1, increases radiosensitization in NSCLC cell lines, as determined by a clonogenic survival assay. Moreover, shUSP14-expressing NSCLC cells show increased NHEJ efficiency, as indicated by chromatin recruitment of key NHEJ proteins, NHEJ reporter assay, and increased IR-induced foci formation by 53BP1 and pS2056-DNA-PKcs. Conversely, shUSP14-expressing NSCLC cells show decreased RPA32 and BRCA1 foci formation, suggesting HR-deficiency. These findings identify USP14 as an important determinant of DSB repair in response to radiotherapy and a promising target for NSCLC radiosensitization.
Collapse
Affiliation(s)
- Arishya Sharma
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Alexandru Almasan
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
121
|
Qin N, Wang Z, Liu Q, Song N, Wilson CL, Ehrhardt MJ, Shelton K, Easton J, Mulder H, Kennetz D, Edmonson MN, Rusch MC, Downing JR, Hudson MM, Nichols KE, Zhang J, Robison LL, Yasui Y. Pathogenic Germline Mutations in DNA Repair Genes in Combination With Cancer Treatment Exposures and Risk of Subsequent Neoplasms Among Long-Term Survivors of Childhood Cancer. J Clin Oncol 2020; 38:2728-2740. [PMID: 32496904 PMCID: PMC7430217 DOI: 10.1200/jco.19.02760] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE To investigate cancer treatment plus pathogenic germline mutations (PGMs) in DNA repair genes (DRGs) for identification of childhood cancer survivors at increased risk of subsequent neoplasms (SNs). METHODS Whole-genome sequencing was performed on blood-derived DNA from survivors in the St Jude Lifetime Cohort. PGMs were evaluated in 127 genes from 6 major DNA repair pathways. Cumulative doses of chemotherapy and body region-specific radiotherapy (RT) were abstracted from medical records. Relative rates (RRs) and 95% CIs of SNs by mutation status were estimated using multivariable piecewise exponential models. RESULTS Of 4,402 survivors, 495 (11.2%) developed 1,269 SNs. We identified 538 PGMs in 98 DRGs (POLG, MUTYH, ERCC2, and BRCA2, among others) in 508 (11.5%) survivors. Mutations in homologous recombination (HR) genes were significantly associated with an increased rate of subsequent female breast cancer (RR, 3.7; 95% CI, 1.8 to 7.7), especially among survivors with chest RT ≥ 20 Gy (RR, 4.4; 95% CI, 1.6 to 12.4), or with a cumulative dose of anthracyclines in the second or third tertile (RR, 4.4; 95% CI, 1.7 to 11.4). Mutations in HR genes were also associated with an increased rate of subsequent sarcoma among those who received alkylating agent doses in the third tertile (RR, 14.9; 95% CI, 4.0 to 38.0). Mutations in nucleotide excision repair genes were associated with subsequent thyroid cancer for those treated with neck RT ≥ 30 Gy (RR, 12.9; 95% CI, 1.6 to 46.6) with marginal statistical significance. CONCLUSION Our study provides novel insights regarding the contribution of genetics, in combination with known treatment-related risks, for the development of SNs. These findings have the potential to facilitate identification of high-risk survivors who may benefit from genetic counseling and/or testing of DRGs, which may further inform personalized cancer surveillance and prevention strategies.
Collapse
Affiliation(s)
- Na Qin
- Department of Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, TN
| | - Zhaoming Wang
- Department of Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, TN
- Department of Computational Biology, St Jude Children’s Research Hospital, Memphis, TN
| | - Qi Liu
- School of Public Health, University of Alberta, Edmonton, Alberta, Canada
| | - Nan Song
- Department of Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, TN
| | - Carmen L. Wilson
- Department of Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, TN
| | - Matthew J. Ehrhardt
- Department of Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, TN
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN
| | - Kyla Shelton
- Department of Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, TN
| | - John Easton
- Department of Computational Biology, St Jude Children’s Research Hospital, Memphis, TN
| | - Heather Mulder
- Department of Computational Biology, St Jude Children’s Research Hospital, Memphis, TN
| | - Dennis Kennetz
- Department of Computational Biology, St Jude Children’s Research Hospital, Memphis, TN
| | - Michael N. Edmonson
- Department of Computational Biology, St Jude Children’s Research Hospital, Memphis, TN
| | - Michael C. Rusch
- Department of Computational Biology, St Jude Children’s Research Hospital, Memphis, TN
| | - James R. Downing
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN
| | - Melissa M. Hudson
- Department of Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, TN
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN
| | - Kim E. Nichols
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN
| | - Jinghui Zhang
- Department of Computational Biology, St Jude Children’s Research Hospital, Memphis, TN
| | - Leslie L. Robison
- Department of Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, TN
| | - Yutaka Yasui
- Department of Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
122
|
Pasculli B, Barbano R, Fontana A, Biagini T, Di Viesti MP, Rendina M, Valori VM, Morritti M, Bravaccini S, Ravaioli S, Maiello E, Graziano P, Murgo R, Copetti M, Mazza T, Fazio VM, Esteller M, Parrella P. Hsa-miR-155-5p Up-Regulation in Breast Cancer and Its Relevance for Treatment With Poly[ADP-Ribose] Polymerase 1 (PARP-1) Inhibitors. Front Oncol 2020; 10:1415. [PMID: 32903519 PMCID: PMC7435065 DOI: 10.3389/fonc.2020.01415] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/06/2020] [Indexed: 01/12/2023] Open
Abstract
miR-155-5p is a well-known oncogenic microRNA, showing frequent overexpression in human malignancies, including breast cancer. Here, we show that high miR-155-5p levels are associated with unfavorable prognostic factors in two independent breast cancer cohorts (CSS cohort, n = 283; and TCGA-BRCA dataset, n = 1,095). Consistently, miR-155-5p results as differentially expressed in the breast cancer subgroups identified by the surrogate molecular classification in the CSS cohort and the PAM50 classifier in TCGA-BRCA dataset, with the TNBC and HER2-amplified tumors carrying the highest levels. Since the analysis of TCGA-BC dataset also demonstrated a significant association between miR-155-5p levels and the presence of mutations in homologous recombination (HR) genes, we hypothesized that miR-155-5p might affect cell response to the PARP-1 inhibitor Olaparib. As expected, miR-155-5p ectopic overexpression followed by Olaparib administration resulted in a greater reduction of cell viability as compared to Olaparib administration alone, suggesting that miR-155-5p might induce a synthetic lethal effect in cancer cells when coupled with PARP-1-inhibition. Overall, our data point to a role of miR-155-5p in homologous recombination deficiency and suggest miR-155-5p might be useful in predicting response to PARP1 inhibitors in the clinical setting.
Collapse
Affiliation(s)
- Barbara Pasculli
- Fondazione IRCCS Casa Sollievo Della Sofferenza, Laboratorio di Oncologia, San Giovanni Rotondo, Italy
| | - Raffaela Barbano
- Fondazione IRCCS Casa Sollievo Della Sofferenza, Laboratorio di Oncologia, San Giovanni Rotondo, Italy
| | - Andrea Fontana
- Fondazione IRCCS Casa Sollievo Della Sofferenza, UO di Biostatistica, San Giovanni Rotondo, Italy
| | - Tommaso Biagini
- Fondazione IRCCS Casa Sollievo Della Sofferenza, Laboratory of Bioinformatics Unit, San Giovanni Rotondo, Italy
| | - Maria Pia Di Viesti
- Fondazione IRCCS Casa Sollievo Della Sofferenza, Laboratorio di Oncologia, San Giovanni Rotondo, Italy
| | - Michelina Rendina
- Fondazione IRCCS Casa Sollievo Della Sofferenza, Laboratorio di Oncologia, San Giovanni Rotondo, Italy
| | - Vanna Maria Valori
- Fondazione IRCCS Casa Sollievo Della Sofferenza, UO di Oncologia, San Giovanni Rotondo, Italy
| | - Maria Morritti
- Fondazione IRCCS Casa Sollievo Della Sofferenza, UO di Oncologia, San Giovanni Rotondo, Italy
| | - Sara Bravaccini
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Biosciences Laboratory, Meldola, Italy
| | - Sara Ravaioli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Biosciences Laboratory, Meldola, Italy
| | - Evaristo Maiello
- Fondazione IRCCS Casa Sollievo Della Sofferenza, UO di Oncologia, San Giovanni Rotondo, Italy
| | - Paolo Graziano
- Fondazione IRCCS Casa Sollievo Della Sofferenza, UO di Anatomia Patologica, San Giovanni Rotondo, Italy
| | - Roberto Murgo
- Fondazione IRCCS Casa Sollievo Della Sofferenza, UO di Chirurgia Senologica, San Giovanni Rotondo, Italy
| | - Massimiliano Copetti
- Fondazione IRCCS Casa Sollievo Della Sofferenza, UO di Biostatistica, San Giovanni Rotondo, Italy
| | - Tommaso Mazza
- Fondazione IRCCS Casa Sollievo Della Sofferenza, Laboratory of Bioinformatics Unit, San Giovanni Rotondo, Italy
| | - Vito Michele Fazio
- Fondazione IRCCS Casa Sollievo Della Sofferenza, Laboratorio di Oncologia, San Giovanni Rotondo, Italy
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain.,Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain.,Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Catalonia, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
| | - Paola Parrella
- Fondazione IRCCS Casa Sollievo Della Sofferenza, Laboratorio di Oncologia, San Giovanni Rotondo, Italy
| |
Collapse
|
123
|
Beca F, Sebastiao APM, Pareja F, Dessources K, Lozada JR, Geyer F, Selenica P, Zeizafoun N, Wen HY, Norton L, Brogi E, Weigelt B, Reis-Filho JS. Whole-exome analysis of metaplastic breast carcinomas with extensive osseous differentiation. Histopathology 2020; 77:321-326. [PMID: 32043609 PMCID: PMC7518240 DOI: 10.1111/his.14088] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/08/2020] [Indexed: 01/08/2023]
Abstract
AIMS Metaplastic breast carcinoma (MBC) is a rare type of triple-negative breast cancer that shows vast histological and genetic heterogeneity. Osseous differentiation can be found in different subtypes of MBC. Whether MBCs with osseous differentiation are underpinned by specific genetic alterations has yet to be defined. The aim of this study was to investigate the repertoire of somatic mutations and copy number alterations (CNAs) in three MBCs with extensive osseous differentiation. METHODS AND RESULTS Tumour and normal DNA samples from three MBCs with extensive osseous differentiation were subjected to whole-exome sequencing. Somatic mutations, CNAs and mutational signatures were determined by use of a validated bioinformatics pipeline. Our analyses revealed clonal TP53 hotspot mutations associated with loss of heterozygosity of the wild-type allele coupled with mutations affecting genes related to the Wnt and/or the phosphoinositide 3-kinase-AKT-mammalian target of rapamycin pathways in all cases analysed. All cases showed a dominant mutational signature 1, with two cases showing a secondary signature 3 in addition to other features of homologous recombination DNA repair defects. The oncostatin M receptor gene, which plays a role in mesenchymal differentiation and bone formation, was found to be mutated in two MBCs with extensive osseous differentiation and in none of 35 previously published 35 MBCs. CONCLUSION Our findings suggest that MBCs with osseous differentiation have somatic mutations similar to those of other forms of MBC.
Collapse
Affiliation(s)
- Francisco Beca
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ana P M Sebastiao
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Positivo University Medical School, Curitiba, Brazil
| | - Fresia Pareja
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kimberly Dessources
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John R Lozada
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Felipe Geyer
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nebras Zeizafoun
- Department of Pathology and Laboratory Medicine, Mount Sinai Healthcare System, New York, NY, USA
| | - Hannah Y Wen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Larry Norton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Edi Brogi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
124
|
Park W, Chen J, Chou JF, Varghese AM, Yu KH, Wong W, Capanu M, Balachandran V, McIntyre CA, El Dika I, Khalil DN, Harding JJ, Ghalehsari N, McKinnell Z, Chalasani SB, Makarov V, Selenica P, Pei X, Lecomte N, Kelsen DP, Abou-Alfa GK, Robson ME, Zhang L, Berger MF, Schultz N, Chan TA, Powell SN, Reis-Filho JS, Iacobuzio-Donahue CA, Riaz N, O'Reilly EM. Genomic Methods Identify Homologous Recombination Deficiency in Pancreas Adenocarcinoma and Optimize Treatment Selection. Clin Cancer Res 2020; 26:3239-3247. [PMID: 32444418 PMCID: PMC7380542 DOI: 10.1158/1078-0432.ccr-20-0418] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/07/2020] [Accepted: 04/08/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Genomic methods can identify homologous recombination deficiency (HRD). Rigorous evaluation of their outcome association to DNA damage response-targeted therapies like platinum in pancreatic ductal adenocarcinoma (PDAC) is essential in maximizing therapeutic outcome. EXPERIMENTAL DESIGN We evaluated progression-free survival (PFS) and overall survival (OS) of patients with advanced-stage PDAC, who had both germline- and somatic-targeted gene sequencing. Homologous recombination gene mutations (HRm) were evaluated: BRCA1, BRCA2, PALB2, ATM, BAP1, BARD1, BLM, BRIP1, CHEK2, FAM175A, FANCA, FANCC, NBN, RAD50, RAD51, RAD51C, and RTEL1 HRm status was grouped as: (i) germline versus somatic; (ii) core (BRCAs and PALB2) versus non-core (other HRm); and (iii) monoallelic versus biallelic. Genomic instability was compared using large-scale state transition, signature 3, and tumor mutation burden. RESULTS Among 262 patients, 50 (19%) had HRD (15% germline and 4% somatic). Both groups were analyzed together due to lack of difference in their genomic instability and outcome. Median [95% confidence interval (CI)] follow-up was 21.9 (1.4-57.0) months. Median OS and PFS were 15.5 (14.6-19) and 7 (6.1-8.1) months, respectively. Patients with HRD had improved PFS compared with no HRD when treated with first-line (1L) platinum [HR, 0.44 (95% CI: 0.29-0.67); P < 0.01], but not with 1L-non-platinum. Multivariate analysis showed HRD patients had improved OS regardless of their first-line treatment, but most had platinum exposure during their course. Biallelic HRm (11%) and core HRm (12%) had higher genomic instability, which translated to improved PFS on first-line platinum (1L-platinum) versus 1L-non-platinum. CONCLUSIONS Pathogenic HRm identifies HRD in patients with PDAC with the best outcome when treated with 1L-platinum. Biallelic HRm and core HRm further enriched benefit from 1L-platinum from HRD.
Collapse
Affiliation(s)
- Wungki Park
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Gastrointestinal Oncology Service, Weill Cornell Medical College, New York, New York
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Jiapeng Chen
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joanne F Chou
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology and Biostatistics, Weill Cornell Medical College, New York, New York
| | - Anna M Varghese
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Gastrointestinal Oncology Service, Weill Cornell Medical College, New York, New York
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kenneth H Yu
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Gastrointestinal Oncology Service, Weill Cornell Medical College, New York, New York
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Winston Wong
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marinela Capanu
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology and Biostatistics, Weill Cornell Medical College, New York, New York
| | - Vinod Balachandran
- Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
- Parker Institute for Cancer Immunotherapy, San Francisco, California
- Department of Surgery, Hepatopancreaticobiliary Surgery, Weill Cornell Medical College, New York, New York
| | - Caitlin A McIntyre
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Surgery, Hepatopancreaticobiliary Surgery, Weill Cornell Medical College, New York, New York
| | - Imane El Dika
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Gastrointestinal Oncology Service, Weill Cornell Medical College, New York, New York
| | - Danny N Khalil
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Gastrointestinal Oncology Service, Weill Cornell Medical College, New York, New York
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - James J Harding
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Gastrointestinal Oncology Service, Weill Cornell Medical College, New York, New York
| | | | - Zoe McKinnell
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sree B Chalasani
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Gastrointestinal Oncology Service, Weill Cornell Medical College, New York, New York
| | - Vladimir Makarov
- Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pier Selenica
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pathology, Weill Cornell Medical College, New York, New York
| | - Xin Pei
- Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nicolas Lecomte
- Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pathology, Weill Cornell Medical College, New York, New York
| | - David P Kelsen
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Gastrointestinal Oncology Service, Weill Cornell Medical College, New York, New York
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ghassan K Abou-Alfa
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Gastrointestinal Oncology Service, Weill Cornell Medical College, New York, New York
| | - Mark E Robson
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Clinical Genetics Service, Weill Cornell Medical College, New York, New York
| | - Liying Zhang
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Gastrointestinal Oncology Service, Weill Cornell Medical College, New York, New York
- Department of Pathology, Weill Cornell Medical College, New York, New York
| | - Michael F Berger
- Memorial Sloan Kettering Cancer Center, New York, New York
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nikolaus Schultz
- Memorial Sloan Kettering Cancer Center, New York, New York
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Timothy A Chan
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Simon N Powell
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
| | - Jorge S Reis-Filho
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pathology, Weill Cornell Medical College, New York, New York
| | - Christine A Iacobuzio-Donahue
- Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pathology, Weill Cornell Medical College, New York, New York
| | - Nadeem Riaz
- Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eileen M O'Reilly
- Memorial Sloan Kettering Cancer Center, New York, New York.
- Department of Medicine, Gastrointestinal Oncology Service, Weill Cornell Medical College, New York, New York
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
125
|
Liu YL, Selenica P, Zhou Q, Iasonos A, Callahan M, Feit NZ, Boland J, Vazquez-Garcia I, Mandelker D, Zehir A, Burger RA, Powell DJ, Friedman C, Cadoo K, Grisham R, Konner JA, O'Cearbhaill RE, Aghajanian C, Reis-Filho JS, Weigelt B, Zamarin D. BRCA Mutations, Homologous DNA Repair Deficiency, Tumor Mutational Burden, and Response to Immune Checkpoint Inhibition in Recurrent Ovarian Cancer. JCO Precis Oncol 2020; 4:2000069. [PMID: 32923884 DOI: 10.1200/po.20.00069] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2020] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Homologous DNA repair-deficient (HRD) ovarian cancers (OCs), including those with BRCA1/2 mutations, have higher levels of genetic instability, potentially resulting in higher immunogenicity, and have been suggested to respond better to immune checkpoint inhibitors (ICIs) than homologous DNA repair-proficient OCs. However, clinical evidence is lacking. The study aimed to evaluate the associations between BRCA1/2 mutations, HRD, and other genomic parameters and response to ICIs and survival in OC. METHODS This is a single-institution retrospective analysis of women with recurrent OC treated with ICIs. BRCA1/2 mutation status and clinicopathologic variables were abstracted from the medical records. Targeted and whole-exome sequencing data available for a subset of patients were used to assess tumor mutational burden (TMB), HRD, and fraction of genome altered (FGA). ICI response was defined as lack of disease progression for ≥ 24 weeks. Associations of BRCA1/2 status and genomic alterations with progression-free survival (PFS) and overall survival (OS) were determined using Cox proportional hazards models. RESULTS Of the 143 women treated with ICIs, 134 had known BRCA1/2 mutation status. Deleterious germline or somatic BRCA1/2 mutations were present in 31 women (24%). There was no association between presence of BRCA1/2 mutations and response (P = .796) or survival. Genomic analysis in 73 women found no association between TMB (P = .344) or HRD (P = .222) and response, PFS, or OS. There were also no significant differences in somatic genetic alterations between responders and nonresponders. High FGA was associated with an improvement in PFS (P = .014) and OS (P = .01). CONCLUSION TMB, BRCA1/2 mutations, and HRD are not associated with response or survival, cautioning against their use as selection criteria for ICI in recurrent OC. FGA should be investigated further as a biomarker of response to immunotherapy in OC.
Collapse
Affiliation(s)
- Ying L Liu
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Qin Zhou
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexia Iasonos
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Margaret Callahan
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY.,Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Noah Z Feit
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Julia Boland
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Diana Mandelker
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ahmet Zehir
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert A Burger
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA
| | - Daniel J Powell
- Department of Pathology, University of Pennsylvania, Philadelphia, PA
| | - Claire Friedman
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Karen Cadoo
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Rachel Grisham
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Jason A Konner
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Roisin E O'Cearbhaill
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Carol Aghajanian
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Dmitriy Zamarin
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY.,Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
126
|
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: 3.4] [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
|
127
|
Paluch-Shimon S, Cardoso F, Partridge AH, Abulkhair O, Azim HA, Bianchi-Micheli G, Cardoso MJ, Curigliano G, Gelmon KA, Harbeck N, Merschdorf J, Poortmans P, Pruneri G, Senkus E, Spanic T, Stearns V, Wengström Y, Peccatori F, Pagani O. ESO-ESMO 4th International Consensus Guidelines for Breast Cancer in Young Women (BCY4). Ann Oncol 2020; 31:674-696. [PMID: 32199930 DOI: 10.1016/j.annonc.2020.03.284] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/07/2020] [Indexed: 12/16/2022] Open
Abstract
The 4th International Consensus Conference for Breast Cancer in Young Women (BCY4) took place in October 2018, in Lugano, Switzerland, organized by the European School of Oncology (ESO) and the European Society of Medical Oncology (ESMO). Consensus recommendations for the management of breast cancer in young women were updated from BCY3 with incorporation of new evidence to inform the guidelines. Areas of research priorities were also identified. This article summarizes the ESO-ESMO international consensus recommendations, which are also endorsed by the European Society of Breast Specialists (EUSOMA).
Collapse
Affiliation(s)
| | - F Cardoso
- Breast Unit Champalimaud Clinical Centre/Champalimaud Foundation, Lisbon, Portugal
| | - A H Partridge
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - O Abulkhair
- King Abdulaziz Medical City for National Guard, Riyadh, Saudi Arabia
| | - H A Azim
- School of Medicine, Monterrey Institute of Technology, Monterrey, MX
| | | | - M-J Cardoso
- Breast Unit Champalimaud Clinical Centre/Champalimaud Foundation, Lisbon, Portugal; Nova Medical School Lisbon, Portugal
| | - G Curigliano
- European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - K A Gelmon
- British Columbia Cancer, Vancouver, Canada
| | - N Harbeck
- Breast Center, Dept. OB&GYN, University of Munich (LMU), Munich, Germany
| | | | - P Poortmans
- Institut Curie, Department of Radiation Oncology & Paris Sciences & Lettres - PSL University, Paris, France
| | - G Pruneri
- National Cancer Institute, IRCCS Foundation, Milan, Italy
| | - E Senkus
- Medical University of Gdansk, Gdansk, Poland
| | - T Spanic
- Europa Donna Slovenia, Ljubljana, Slovenia
| | - V Stearns
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, USA
| | - Y Wengström
- Department of Neurobiology Cancer Science and Society, Karolinska Institute and Theme Cancer Karolinska University Hospital, Sweden
| | - F Peccatori
- European Institute of Oncology IRCCS & European School of Oncology, Milan, Italy
| | - O Pagani
- Oncology Institute of Southern Switzerland and Breast Unit of Southern Switzerland, Geneva University Hospitals, Swiss Group for Clinical Cancer Research (SAKK), Bellinzona, Switzerland
| |
Collapse
|
128
|
Beddok A, Krieger S, Castera L, Stoppa-Lyonnet D, Thariat J. Management of Fanconi Anemia patients with head and neck carcinoma: Diagnosis and treatment adaptation. Oral Oncol 2020; 108:104816. [PMID: 32480311 DOI: 10.1016/j.oraloncology.2020.104816] [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: 11/09/2019] [Accepted: 05/20/2020] [Indexed: 12/18/2022]
Abstract
Fanconi anemia (FA) is a rare genetic disease that is mostly transmitted, according to a recessive model with biallelic germline alterations in one of the 22 genes of the FA pathway, or monoallelic alteration of the 23rd FA gene (RAD51). The FA pathway is implicated in interstrand DNA crosslink repair, induces genome stability, and is a potent driver of tumorigenesis. Patients with FA have a 500 to 1000-fold increased risk of developing head and neck squamous cell carcinoma (HNSCC). Patients with FA developing an HNSCC, usually have severe radiation toxicities. In this context, the modalities of radiation therapy should be adapted. Some patients with FA present a milder phenotype, especially in the case of medullary FA gene spontaneous reversion. Therefore, in an unusual context of HNSCC, such as no risk factors or a young age, it may be very useful to search anemia or development abnormalities, that may unravel a yet undiagnosed FA disease. Besides, in some young patients with HNSCC who did not suffer from FA, a monoallelic germline alteration in an FA gene could be combined with a second risk factor such as HPV infection or APOBEC alteration. Although several in vitro studies showed that normal cells with monoallelic FA gene alteration may have a particular radiosensitivity, these observations have not been confirmed in vivo in FA heterozygotes patients. Finally, some somatic activating alterations have also been found in HSNCC tumor samples and could be associated with radioresistance.
Collapse
Affiliation(s)
- Arnaud Beddok
- Department of Radiation Oncology, Curie Institute, Paris, France.
| | - Sophie Krieger
- Department of Cancer Biology and Genetics, Inserm U1245, Normandy Centre for Genomic and Personalized Medicine, François Baclesse Center, Caen, France
| | - Laurent Castera
- Department of Cancer Biology and Genetics, Inserm U1245, Normandy Centre for Genomic and Personalized Medicine, François Baclesse Center, Caen, France
| | | | - Juliette Thariat
- Department of Radiation Oncology, François Baclesse Center, Caen, France
| |
Collapse
|
129
|
Santana dos Santos E, Lallemand F, Petitalot A, Caputo SM, Rouleau E. HRness in Breast and Ovarian Cancers. Int J Mol Sci 2020; 21:E3850. [PMID: 32481735 PMCID: PMC7312125 DOI: 10.3390/ijms21113850] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/25/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023] Open
Abstract
Ovarian and breast cancers are currently defined by the main pathways involved in the tumorigenesis. The majority are carcinomas, originating from epithelial cells that are in constant division and subjected to cyclical variations of the estrogen stimulus during the female hormonal cycle, therefore being vulnerable to DNA damage. A portion of breast and ovarian carcinomas arises in the context of DNA repair defects, in which genetic instability is the backdrop for cancer initiation and progression. For these tumors, DNA repair deficiency is now increasingly recognized as a target for therapeutics. In hereditary breast/ovarian cancers (HBOC), tumors with BRCA1/2 mutations present an impairment of DNA repair by homologous recombination (HR). For many years, BRCA1/2 mutations were only screened on germline DNA, but now they are also searched at the tumor level to personalize treatment. The reason of the inactivation of this pathway remains uncertain for most cases, even in the presence of a HR-deficient signature. Evidence indicates that identifying the mechanism of HR inactivation should improve both genetic counseling and therapeutic response, since they can be useful as new biomarkers of response.
Collapse
Affiliation(s)
- Elizabeth Santana dos Santos
- Department of Medical Biology and Pathology, Gustave Roussy, Cancer Genetics Laboratory, Gustave Roussy, 94800 Villejuif, France;
- Department of Clinical Oncology, A.C. Camargo Cancer Center, São Paulo 01509-010, Brazil
| | - François Lallemand
- Department of Genetics, Institut Curie, 75005 Paris, France; (F.L.); (A.P.); (S.M.C.)
- PSL Research University, 75005 Paris, France
| | - Ambre Petitalot
- Department of Genetics, Institut Curie, 75005 Paris, France; (F.L.); (A.P.); (S.M.C.)
- PSL Research University, 75005 Paris, France
| | - Sandrine M. Caputo
- Department of Genetics, Institut Curie, 75005 Paris, France; (F.L.); (A.P.); (S.M.C.)
- PSL Research University, 75005 Paris, France
| | - Etienne Rouleau
- Department of Medical Biology and Pathology, Gustave Roussy, Cancer Genetics Laboratory, Gustave Roussy, 94800 Villejuif, France;
| |
Collapse
|
130
|
Sokol ES, Pavlick D, Khiabanian H, Frampton GM, Ross JS, Gregg JP, Lara PN, Oesterreich S, Agarwal N, Necchi A, Miller VA, Alexander B, Ali SM, Ganesan S, Chung JH. Pan-Cancer Analysis of BRCA1 and BRCA2 Genomic Alterations and Their Association With Genomic Instability as Measured by Genome-Wide Loss of Heterozygosity. JCO Precis Oncol 2020; 4:442-465. [PMID: 32903788 PMCID: PMC7446440 DOI: 10.1200/po.19.00345] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2020] [Indexed: 12/27/2022] Open
Abstract
PURPOSE BRCA1 or BRCA2 loss of function results in homologous recombination deficiency (HRD), which is targetable by poly (ADP-ribose) polymerase (PARP) inhibitors and other DNA-damaging agents. In cancers associated with germline BRCA1/2 alterations (BRCA1/2-associated cancers: breast, ovarian, pancreatic, prostate), BRCA1/2 alterations result in HRD and are biomarkers for PARP inhibitor use. In other (non-BRCA1/2-associated) cancer types, the association between BRCA1/2 alteration and HRD is less clear. METHODS A total of 234,154 tumor samples were sequenced by hybrid capture-based comprehensive genomic profiling. Somatic, germline, and zygosity status was determined computationally. BRCA1/2 alterations were classified as predicted germline/somatic and biallelic/monoallelic. Genome-wide loss of heterozygosity (gLOH) was evaluated as a marker of HRD. RESULTS BRCA1/2 alterations were observed at a 4.7% frequency. BRCA1/2 mutations were predicted germline in 57.4% of BRCA1/2-associated and 37.2% of non-BRCA1/2-associated cancers. The fraction of BRCA1/2-altered cases that were biallelic was 68.7%, with a higher biallelic fraction in BRCA1/2-associated (89.9%) versus non-BRCA1/2-associated cancers (43.6%). Differences in tissue distribution of biallelic BRCA1 versus BRCA2 alterations were noted, including a higher rate of biallelic BRCA2 alteration in prostate cancer. Biallelic BRCA1/2 alteration was observed at a 3.2% frequency (BRCA1/2-associated cancers, 8.9%; non-BRCA1/2-associated cancers, 1.3%) and > 1% frequency in at least 13 cancer types. Across cancer types, biallelic BRCA1/2 alteration was associated with increased gLOH versus monoallelic or wild-type BRCA1/2; predicted germline or somatic mutations were both associated with elevated gLOH. CONCLUSION Biallelic BRCA1/2 alterations were associated with elevated gLOH in diverse cancer types, including those not traditionally associated with BRCA1/2 cancer syndromes. Biomarker development for PARP inhibitors should integrate methods to distinguish biallelic from monoallelic BRCA1/2 status, and biallelic BRCA1/2 alteration should be broadly evaluated across cancer types as a biomarker for underlying HRD and PARP inhibitor sensitivity.
Collapse
Affiliation(s)
| | | | - Hossein Khiabanian
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ
| | | | - Jeffrey S. Ross
- Foundation Medicine, Cambridge, MA
- Upstate Medical University, Syracuse, NY
| | | | - Primo N. Lara
- University of California Davis Medical Center, Sacramento, CA
| | - Steffi Oesterreich
- University of Pittsburgh, Womens Cancer Research Center/Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, Pittsburgh, PA
| | | | - Andrea Necchi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | | | - Shridar Ganesan
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ
| | | |
Collapse
|
131
|
Points to consider for reporting of germline variation in patients undergoing tumor testing: a statement of the American College of Medical Genetics and Genomics (ACMG). Genet Med 2020; 22:1142-1148. [PMID: 32321997 DOI: 10.1038/s41436-020-0783-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 02/08/2023] Open
|
132
|
Condorelli R, Mosele F, Verret B, Bachelot T, Bedard PL, Cortes J, Hyman DM, Juric D, Krop I, Bieche I, Saura C, Sotiriou C, Cardoso F, Loibl S, Andre F, Turner NC. Genomic alterations in breast cancer: level of evidence for actionability according to ESMO Scale for Clinical Actionability of molecular Targets (ESCAT). Ann Oncol 2020; 30:365-373. [PMID: 30715161 DOI: 10.1093/annonc/mdz036] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Better knowledge of the tumor genomic landscapes has helped to develop more effective targeted drugs. However, there is no tool to interpret targetability of genomic alterations assessed by next-generation sequencing in the context of clinical practice. Our aim is to rank the level of evidence of individual recurrent genomic alterations observed in breast cancer based on the ESMO Scale for Clinical Actionability of molecular Targets (ESCAT) in order to help the clinicians to prioritize treatment. Analyses of databases suggested that there are around 40 recurrent driver alterations in breast cancer. ERBB2 amplification, germline BRCA1/2 mutations, PIK3CA mutations were classified tier of evidence IA based on large randomized trials showing antitumor activity of targeted therapies in patients presenting the alterations. NTRK fusions and microsatellite instability (MSI) were ranked IC. ESR1 mutations and PTEN loss were ranked tier IIA, and ERBB2 mutations and AKT1 mutations tier IIB. Somatic BRCA 1/2 mutations, MDM2 amplifications and ERBB 3 mutations were ranked tier III. Seventeen genes were ranked tier IV based on preclinical evidence. Finally, FGFR1 and CCND1 were ranked tier X alterations because previous studies have shown lack of actionability.
Collapse
Affiliation(s)
- R Condorelli
- Department of Medical Oncolo, INSERM U981, Université Paris Sud, Gustave Roussy, Villejuif, France; Institute of Oncology and Breast Unit of Southern Switzerland, Bellinzona, Switzerland
| | - F Mosele
- Department of Medical Oncolo, INSERM U981, Université Paris Sud, Gustave Roussy, Villejuif, France.
| | - B Verret
- Department of Medical Oncolo, INSERM U981, Université Paris Sud, Gustave Roussy, Villejuif, France
| | - T Bachelot
- Department of Medical Oncology, Cancer Research Center of Lyon Inserm, Lyon, France
| | - P L Bedard
- Division of Medical Oncology & Hematolog, Department of Medicine, Princess Margaret Cancer Centre, Toronto, Canada
| | - J Cortes
- Ramon y Cajal University Hospital, Madrid & Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - D M Hyman
- Memorial Sloan Kettering Cancer Center, New York
| | - D Juric
- Massachusetts General Hospital (MGH), Boston
| | - I Krop
- Dana-Farber Cancer Institute, Boston, USA
| | - I Bieche
- Department of Genetics, Curie Institute, Paris, France
| | - C Saura
- Department of Medical Oncolog, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - C Sotiriou
- J.C. Heuson Breast Cancer Translational Research Laborator, Université Libre de Bruxelles, Institut Jules Bordet, Brussels, Belgium
| | - F Cardoso
- Breast Uni, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| | - S Loibl
- German Breast Group, Neu-Isenburg, Germany
| | - F Andre
- Department of Medical Oncolo, INSERM U981, Université Paris Sud, Gustave Roussy, Villejuif, France
| | - N C Turner
- Royal Marsden Hospital and Institute of Cancer Research, London, UK
| |
Collapse
|
133
|
Chou J, Quigley DA, Robinson TM, Feng FY, Ashworth A. Transcription-Associated Cyclin-Dependent Kinases as Targets and Biomarkers for Cancer Therapy. Cancer Discov 2020; 10:351-370. [DOI: 10.1158/2159-8290.cd-19-0528] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/29/2019] [Accepted: 11/04/2019] [Indexed: 11/16/2022]
|
134
|
Current concepts in breast cancer genomics: An evidence based review by the CGC breast cancer working group. Cancer Genet 2020; 244:11-20. [PMID: 32087595 DOI: 10.1016/j.cancergen.2020.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 01/18/2020] [Accepted: 02/05/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Genomic abnormalities in breast cancer have been described according to diverse conceptual frameworks, including histologic subtypes, clinical molecular subtypes, intrinsic DNA, RNA, and epigenetic profiles, and activated molecular pathways. METHODS The Cancer Genomics Consortium (CGC) Breast Cancer Workgroup performed an evidence based literature review to summarize current knowledge of clinically significant genomic alterations in breast cancer using CGC levels of evidence. Targetable or disease-defining alterations were prioritized. RESULTS We summarized genomic alterations in breast cancer within a framework of existing clinical tools for diagnosis, risk stratification, and therapeutic management. Using CGC levels of evidence, we catalog copy number profiles, gene expression profiles, and mutations in clinically significant genes. We also describe emerging molecular markers such as methylation profiling and immunotherapy biomarkers. CONCLUSION A summary of currently available information on breast cancer genomics will enhance precision medicine by serving as an interpretive resource for clinical laboratory geneticists, providing a foundation for future practice guidelines, and identifying knowledge gaps to address in future research.
Collapse
|
135
|
Zeng ZM, Du HY, Xiong L, Zeng XL, Zhang P, Cai J, Huang L, Liu AW. BRCA1 protects cardiac microvascular endothelial cells against irradiation by regulating p21-mediated cell cycle arrest. Life Sci 2020; 244:117342. [PMID: 31978450 DOI: 10.1016/j.lfs.2020.117342] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/12/2020] [Accepted: 01/20/2020] [Indexed: 12/14/2022]
Abstract
AIMS Microvascular endothelial cell dysfunction is a leading cause of radiation-induced heart disease (RIHD). BRCA1 plays an important role in DNA damage repair. The study aims to explore the effect of BRCA1 in endothelial cells involved in RIHD. MATERIALS AND METHODS BRCA1 and p21 expression were detected in human umbilical vein endothelial cells (HUVECs) and in mouse heart tissue after irradiation exposure. The effects of BRCA1 on cell proliferation, cell cycle and radiosensitivity were determined in HUVECs with overexpression and knockdown of BRCA1. A mouse model of RIHD was established. Heart damage was detected in C57BL/6J mice and endothelial cell specific knockout BRCA1 mice (EC-BRCA1-/-). KEY FINDINGS BRCA1 and p21 expression was significantly increased both in vitro and vivo response to irradiation. BRCA1 overexpression in endothelial cells enhanced cell growth and G1/S phase arrest, and the opposite results were observed in BRCA1 knockdown endothelial cells. BRCA1 downregulated endothelial cell cycle-related genes cyclin A, cyclin D1, cyclin E and p-Rb through increasing p21 expression, and HUVECs with BRCA1 gene knockdown were more sensitive to radiation. In vivo, a decrease in cardiac microvascular density, as well as cardiomyocyte hypoxia and apoptosis were observed in a time-dependent manner. EC-BRCA1-/- mice were more prone to severe RIHD than EC-BRCA1+/- mice after 16Gy radiation exposure due to endothelial dysfunction caused by loss of BRCA1, and p21 was declined in EC-BRCA1-/- mice heart. SIGNIFICANCE These findings indicate that BRCA1 plays a protective role in RIHD by regulating endothelial cell cycle arrest mediated by p21 signal.
Collapse
Affiliation(s)
- Zhi-Min Zeng
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China
| | - Hai-Yang Du
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China
| | - Le Xiong
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China
| | - Xiao-Li Zeng
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
| | - Peng Zhang
- Department of Radiation Oncology, Zhejiang Key Laboratory of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, PR China
| | - Jing Cai
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
| | - Long Huang
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China.
| | - An-Wen Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China.
| |
Collapse
|
136
|
Next-generation sequencing informs diagnosis and identifies unexpected therapeutic targets in lung squamous cell carcinomas. Lung Cancer 2019; 140:35-41. [PMID: 31855703 DOI: 10.1016/j.lungcan.2019.12.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/08/2019] [Accepted: 12/10/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Potentially targetable genomic alterations have been identified in lung squamous cell carcinoma (LUSC), but none have yet translated into effective therapy. We examined potential benefits of next generation sequencing (NGS) in a cohort of consecutive LUSC patients with emphasis on distinctions between smokers and light/never smokers and implications for clinical trial enrollment. METHODS We retrospectively evaluated results from an internally developed NGS assay (OncoPanel) targeting ∼300 genes with a mean overall target coverage of >200x for consecutive LUSC seen at our institution over 30 months. RESULTS Tissue was obtained from 172 patients for targeted NGS. 42 (24 %) samples were insufficient for testing. Median age of tested patients was 66, including 87 % moderate/heavy versus 13 % light/never smokers; 66 % were stage IIIB or IV. Of 130 patients with evaluable NGS results, 49 (38 %) had at least 1 alteration qualifying for enrollment to a LungMAP treatment arm (PIK3CA, MET, FGFR family, cell cycle, or homologous recombination pathways) or for an approved therapy or other clinical trial (e.g. EGFR sensitizing mutations, MET exon 14 splice mutations, TSC1/2 mutation, or microsatellite instability). Therapeutic targets were enriched in light/never smokers (47 % vs 35 % moderate/heavy smokers). Unexpectedly, genomic features suggested an alternative diagnosis (metastatic cutaneous squamous carcinoma; mesothelioma) in 7 patients, including 35 % of never/light smokers. CONCLUSION NGS in a real-world LUSC cohort yields potentially targetable genomic alterations informing clinical trial enrollment and approved therapies and critical diagnostic insights. Our findings strongly support current guidelines recommending mutational profiling of LUSC arising in light/never smoking patients; the utility of sequencing in smokers with LUSC appears to be limited to identification of research targets.
Collapse
|
137
|
Bergom C, West CM, Higginson DS, Abazeed ME, Arun B, Bentzen SM, Bernstein JL, Evans JD, Gerber NK, Kerns SL, Keen J, Litton JK, Reiner AS, Riaz N, Rosenstein BS, Sawakuchi GO, Shaitelman SF, Powell SN, Woodward WA. The Implications of Genetic Testing on Radiation Therapy Decisions: A Guide for Radiation Oncologists. Int J Radiat Oncol Biol Phys 2019; 105:698-712. [PMID: 31381960 PMCID: PMC11913060 DOI: 10.1016/j.ijrobp.2019.07.026] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 06/21/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023]
Abstract
The advent of affordable and rapid next-generation DNA sequencing technology, along with the US Supreme Court ruling invalidating gene patents, has led to a deluge of germline and tumor genetic variant tests that are being rapidly incorporated into clinical cancer decision-making. A major concern for clinicians is whether the presence of germline mutations may increase the risk of radiation toxicity or secondary malignancies. Because scarce clinical data exist to inform decisions at this time, the American Society for Radiation Oncology convened a group of radiation science experts and clinicians to summarize potential issues, review relevant data, and provide guidance for adult patients and their care teams regarding the impact, if any, that genetic testing should have on radiation therapy recommendations. During the American Society for Radiation Oncology workshop, several main points emerged, which are discussed in this manuscript: (1) variants of uncertain significance should be considered nondeleterious until functional genomic data emerge to demonstrate otherwise; (2) possession of germline alterations in a single copy of a gene critical for radiation damage responses does not necessarily equate to increased risk of radiation-induced toxicity; (3) deleterious ataxia-telangiesctasia gene mutations may modestly increase second cancer risk after radiation therapy, and thus follow-up for these patients after indicated radiation therapy should include second cancer screening; (4) conveying to patients the difference between relative and absolute risk is critical to decision-making; and (5) more work is needed to assess the impact of tumor somatic alterations on the probability of response to radiation therapy and the potential for individualization of radiation doses. Data on radiosensitivity related to specific genetic mutations is also briefly discussed.
Collapse
Affiliation(s)
- Carmen Bergom
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Catharine M West
- Division of Cancer Sciences, National Institute for Health Research Manchester Biomedical Research Centre, University of Manchester, Christie National Health Service Foundation Trust Hospital, Manchester, UK
| | - Daniel S Higginson
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mohamed E Abazeed
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, Ohio; Department of Translational Hematology Oncology Research, Cleveland Clinic, Cleveland, Ohio
| | - Banu Arun
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Soren M Bentzen
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jonine L Bernstein
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jaden D Evans
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology and Precision Genomics, Intermountain Healthcare, Ogden, Utah
| | - Naamit K Gerber
- Department of Radiation Oncology, New York University Langone Health, New York, New York
| | - Sarah L Kerns
- Department of Radiation Oncology, University of Rochester, Rochester, New York
| | - Judy Keen
- Scientific Affairs, American Society for Radiation Oncology, Arlington, Virginia
| | - Jennifer K Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anne S Reiner
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Barry S Rosenstein
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Gabriel O Sawakuchi
- Department of Radiation Physics The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Simona F Shaitelman
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Simon N Powell
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Wendy A Woodward
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
138
|
Póti Á, Gyergyák H, Németh E, Rusz O, Tóth S, Kovácsházi C, Chen D, Szikriszt B, Spisák S, Takeda S, Szakács G, Szallasi Z, Richardson AL, Szüts D. Correlation of homologous recombination deficiency induced mutational signatures with sensitivity to PARP inhibitors and cytotoxic agents. Genome Biol 2019; 20:240. [PMID: 31727117 PMCID: PMC6857305 DOI: 10.1186/s13059-019-1867-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/28/2019] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Homologous recombination (HR) repair deficiency arising from defects in BRCA1 or BRCA2 is associated with characteristic patterns of somatic mutations. In this genetic study, we ask whether inactivating mutations in further genes of the HR pathway or the DNA damage checkpoint also give rise to somatic mutation patterns that can be used for treatment prediction. RESULTS Using whole genome sequencing of an isogenic knockout cell line panel, we find a universal HR deficiency-specific base substitution signature that is similar to COSMIC signature 3. In contrast, we detect different deletion phenotypes corresponding to specific HR mutants. The inactivation of BRCA2 or PALB2 leads to larger deletions, typically with microhomology, when compared to the disruption of BRCA1, RAD51 paralogs, or RAD54. Comparison with the deletion spectrum of Cas9 cut sites suggests that most spontaneously arising genomic deletions are not the consequence of double-strand breaks. Surprisingly, the inactivation of checkpoint kinases ATM and CHK2 has no mutagenic consequences. Analysis of tumor exomes with biallelic inactivating mutations in the investigated genes confirms the validity of the cell line models. We present a comprehensive analysis of sensitivity of the investigated mutants to 13 therapeutic agents for the purpose of correlating genomic mutagenic phenotypes with drug sensitivity. CONCLUSION Our results suggest that no single genomic mutational class shows perfect correlation with sensitivity to common treatments, but the contribution of COSMIC signature 3 to base substitutions, or a combined measure of different features, may be reasonably good at predicting platinum and PARP inhibitor sensitivity.
Collapse
Affiliation(s)
- Ádám Póti
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudosok krt 2, Budapest, H-1117, Hungary
| | - Hella Gyergyák
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudosok krt 2, Budapest, H-1117, Hungary
| | - Eszter Németh
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudosok krt 2, Budapest, H-1117, Hungary
| | - Orsolya Rusz
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudosok krt 2, Budapest, H-1117, Hungary
- Department of Oncotherapy, University of Szeged, Szeged, Hungary
| | - Szilárd Tóth
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudosok krt 2, Budapest, H-1117, Hungary
| | - Csenger Kovácsházi
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudosok krt 2, Budapest, H-1117, Hungary
| | - Dan Chen
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudosok krt 2, Budapest, H-1117, Hungary
| | - Bernadett Szikriszt
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudosok krt 2, Budapest, H-1117, Hungary
| | - Sándor Spisák
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Shunichi Takeda
- Department of Radiation Genetics, Kyoto University Medical School, Kyoto, 606-8501, Japan
| | - Gergely Szakács
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudosok krt 2, Budapest, H-1117, Hungary
- Institute of Cancer Research, Medical University Vienna, Vienna, Austria
| | - Zoltan Szallasi
- Computational Health Informatics Program (CHIP), Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Danish Cancer Society Research Center, Copenhagen, Denmark
- SE-NAP, Brain Metastasis Research Group, 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | | | - Dávid Szüts
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudosok krt 2, Budapest, H-1117, Hungary.
| |
Collapse
|
139
|
Li P, Xu Y, Zhang Q, Li Y, Jia W, Wang X, Xie Z, Liu J, Zhao D, Shao M, Chen S, Mo N, Jiang Z, Li L, Liu R, Huang W, Chang L, Chen S, Li H, Zuo W, Li J, Zhang R, Yang X. Evaluating the role of RAD52 and its interactors as novel potential molecular targets for hepatocellular carcinoma. Cancer Cell Int 2019; 19:279. [PMID: 31719794 PMCID: PMC6836504 DOI: 10.1186/s12935-019-0996-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 10/23/2019] [Indexed: 02/07/2023] Open
Abstract
Background Radiation sensitive 52 (RAD52) is an important protein that mediates DNA repair in tumors. However, little is known about the impact of RAD52 on hepatocellular carcinoma (HCC). We investigated the expression of RAD52 and its values in HCC. Some proteins that might be coordinated with RAD52 in HCC were also analyzed. Methods Global RAD52 mRNA levels in HCC were assessed using The Cancer Genome Atlas (TCGA) database. RAD52 expression was analyzed in 70 HCC tissues and adjacent tissues by quantitative real-time PCR (qRT-PCR), Western blotting and immunohistochemistry. The effect of over-expressed RAD52 in Huh7 HCC cells was investigated. The String database was then used to perform enrichment and functional analysis of RAD52 and its interactome. Cytoscape software was used to create a protein–protein interaction network. Molecular interaction studies with RAD52 and its interactome were performed using the molecular docking tools in Hex8.0.0. Finally, these DNA repair proteins, which interact with RAD52, were also analyzed using the TCGA dataset and were detected by qRT-PCR. Based on the TCGA database, algorithms combining ROC between RAD52 and RAD52 interactors were used to diagnose HCC by binary logistic regression. Results In TCGA, upregulated RAD52 related to gender was obtained in HCC. The area under the receiver operating characteristic curve (AUC) of RAD52 was 0.704. The results of overall survival (OS) and recurrence-free survival (RFS) indicated no difference in the prognosis between patients with high and low RAD52 gene expression. We validated that RAD52 expression was increased at the mRNA and protein levels in Chinese HCC tissues compared with adjacent tissues. Higher RAD52 was associated with older age, without correlation with other clinicopathological factors. In vitro, over-expressed RAD52 significantly promoted the proliferation and migration of Huh7 cells. Furthermore, RAD52 interactors (radiation sensitive 51, RAD51; X-ray repair cross complementing 6, XRCC6; Cofilin, CFL1) were also increased in HCC and participated in some biological processes with RAD52. Protein structure analysis showed that RAD52–RAD51 had the firmest binding structure with the lowest E-total energy (− 1120.5 kcal/mol) among the RAD52–RAD51, RAD52–CFL1, and RAD52–XRCC6 complexes. An algorithm combining ROC between RAD52 and its interactome indicated a greater specificity and sensitivity for HCC screening. Conclusions Overall, our study suggested that RAD52 plays a vital role in HCC pathogenesis and serves as a potential molecular target for HCC diagnosis and treatment. This study’s findings regarding the multigene prediction and diagnosis of HCC are valuable.
Collapse
Affiliation(s)
- Ping Li
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China.,4College & Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi China.,5Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi China
| | - YanZhen Xu
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China.,8Department of Pathophysiology, Guangxi Medical University, Nanning, Guangxi China
| | - Qinle Zhang
- Genetic and Metabolic Central Laboratory, The Maternal and Children Health Hospital of Guangxi, Guangxi, China
| | - Yu Li
- Medical Science Laboratory at Liuzhou Worker's Hospital, Liuzhou, Guangxi China
| | - Wenxian Jia
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China.,6College of Pharmacy, Guangxi Medical University, Nanning, Guangxi China
| | - Xiao Wang
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Zhibin Xie
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China
| | - Jiayi Liu
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China.,4College & Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi China.,5Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi China
| | - Dong Zhao
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China
| | - Mengnan Shao
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China
| | - Suixia Chen
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China.,8Department of Pathophysiology, Guangxi Medical University, Nanning, Guangxi China
| | - Nanfang Mo
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Zhiwen Jiang
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Liuyan Li
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Run Liu
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Wanying Huang
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Li Chang
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Siyu Chen
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Hongtao Li
- 2Scientific Research Center, Guilin Medical University, Guilin, Guangxi China
| | - Wenpu Zuo
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Jiaquan Li
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | | | - Xiaoli Yang
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China.,2Scientific Research Center, Guilin Medical University, Guilin, Guangxi China
| |
Collapse
|
140
|
Bartha Á, Győrffy B. Comprehensive Outline of Whole Exome Sequencing Data Analysis Tools Available in Clinical Oncology. Cancers (Basel) 2019; 11:E1725. [PMID: 31690036 PMCID: PMC6895801 DOI: 10.3390/cancers11111725] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/31/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022] Open
Abstract
Whole exome sequencing (WES) enables the analysis of all protein coding sequences in the human genome. This technology enables the investigation of cancer-related genetic aberrations that are predominantly located in the exonic regions. WES delivers high-throughput results at a reasonable price. Here, we review analysis tools enabling utilization of WES data in clinical and research settings. Technically, WES initially allows the detection of single nucleotide variants (SNVs) and copy number variations (CNVs), and data obtained through these methods can be combined and further utilized. Variant calling algorithms for SNVs range from standalone tools to machine learning-based combined pipelines. Tools for CNV detection compare the number of reads aligned to a dedicated segment. Both SNVs and CNVs help to identify mutations resulting in pharmacologically druggable alterations. The identification of homologous recombination deficiency enables the use of PARP inhibitors. Determining microsatellite instability and tumor mutation burden helps to select patients eligible for immunotherapy. To pave the way for clinical applications, we have to recognize some limitations of WES, including its restricted ability to detect CNVs, low coverage compared to targeted sequencing, and the missing consensus regarding references and minimal application requirements. Recently, Galaxy became the leading platform in non-command line-based WES data processing. The maturation of next-generation sequencing is reinforced by Food and Drug Administration (FDA)-approved methods for cancer screening, detection, and follow-up. WES is on the verge of becoming an affordable and sufficiently evolved technology for everyday clinical use.
Collapse
Affiliation(s)
- Áron Bartha
- Semmelweis University, Department of Bioinformatics and 2nd Department of Pediatrics, H-1094 Budapest, Hungary.
- TTK Cancer Biomarker Research Group, Institute of Enzymology, Magyar tudósokkörútja 2., H-1117 Budapest, Hungary.
| | - Balázs Győrffy
- Semmelweis University, Department of Bioinformatics and 2nd Department of Pediatrics, H-1094 Budapest, Hungary.
- TTK Cancer Biomarker Research Group, Institute of Enzymology, Magyar tudósokkörútja 2., H-1117 Budapest, Hungary.
| |
Collapse
|
141
|
Principe DR, Kamath SD, Munshi HG, Mohindra NA. Metastatic Thymoma Harboring a Deleterious BRCA2 Mutation Derives Durable Clinical Benefit from Olaparib. Oncologist 2019; 25:301-305. [PMID: 32297440 DOI: 10.1634/theoncologist.2019-0393] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/24/2019] [Indexed: 01/23/2023] Open
Abstract
Thymomas comprise a group of rare epithelial neoplasms of the anterior mediastinum. Whereas localized disease carries a favorable prognosis, the majority of patients with metastatic thymomas experience progression or recurrence over a 10-year period. Although targeted therapies have become standard of care in many malignancies, no clinically actionable mutations have consistently been identified in metastatic thymomas. Here, we describe a patient with an aggressive thymoma complicated by extensive pleural metastases. Over a 16-year period, she progressed on multiple treatment regimens. To identify additional treatment options, tissue from a pleural metastasis was sent for next-generation sequencing, revealing mutations in BRCA2, tyrosine kinase 2, and SET domain containing 2. Based on supporting evidence for poly (ADP-ribose) polymerase (PARP) inhibition in other BRCA-mutated tumors, the patient was started on the PARP inhibitor olaparib. She derived significant clinical benefit from treatment, with imaging showing overall stabilization of her disease. Here, we review the genotyping results of her tumor and discuss the functional and clinical significance of the mutations in her cancer as well as implications for managing patients with advanced BRCA-mutant thymomas. KEY POINTS: Targeted therapy has yet to enter the standard clinical management of metastatic thymomas. Patients with BRCA2-mutant thymomas may benefit from poly (ADP-ribose) polymerase inhibition.
Collapse
Affiliation(s)
- Daniel R Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Suneel D Kamath
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Nisha A Mohindra
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
142
|
|
143
|
El-Kenawy A, Benarba B, Neves AF, de Araujo TG, Tan BL, Gouri A. Gene surgery: Potential applications for human diseases. EXCLI JOURNAL 2019; 18:908-930. [PMID: 31762718 PMCID: PMC6868916 DOI: 10.17179/excli2019-1833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022]
Abstract
Gene therapy became in last decade a new emerging therapeutic era showing promising results against different diseases such as cancer, cardiovascular diseases, diabetes, and neurological disorders. Recently, the genome editing technique for eukaryotic cells called CRISPR-Cas (Clustered Regulatory Interspaced Short Palindromic Repeats) has enriched the field of gene surgery with enhanced applications. In the present review, we summarized the different applications of gene surgery for treating human diseases such as cancer, diabetes, nervous, and cardiovascular diseases, besides the molecular mechanisms involved in these important effects. Several studies support the important therapeutic applications of gene surgery in a large number of health disorders and diseases including β-thalassemia, cancer, immunodeficiencies, diabetes, and neurological disorders. In diabetes, gene surgery was shown to be effective in type 1 diabetes by triggering different signaling pathways. Furthermore, gene surgery, especially that using CRISPR-Cas possessed important application on diagnosis, screening and treatment of several cancers such as lung, liver, pancreatic and colorectal cancer. Nevertheless, gene surgery still presents some limitations such as the design difficulties and costs regarding ZFNs (Zinc Finger Nucleases) and TALENs (Transcription Activator-Like Effector Nucleases) use, off-target effects, low transfection efficiency, in vivo delivery-safety and ethical issues.
Collapse
Affiliation(s)
- Ayman El-Kenawy
- Department of Pathology, College of Medicine, Taif University, Saudi Arabia
- Department of Molecular Biology, GEBRI, University of Sadat City, P.O. Box 79, Sadat City, Egypt
| | - Bachir Benarba
- Laboratory Research on Biological Systems and Geomatics, Faculty of Nature and Life Sciences, University of Mascara, Algeria
| | - Adriana Freitas Neves
- Institute of Biotechnology, Molecular Biology Laboratory, Universidade Federal de Goias, Catalao, Brazil
| | - Thaise Gonçalves de Araujo
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas, MG, Brazil
| | - Bee Ling Tan
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Adel Gouri
- Laboratory of Medical Biochemistry, Faculty of Medicine, University of Annaba, Algeria
| |
Collapse
|
144
|
Urbina-Jara LK, Rojas-Martinez A, Martinez-Ledesma E, Aguilar D, Villarreal-Garza C, Ortiz-Lopez R. Landscape of Germline Mutations in DNA Repair Genes for Breast Cancer in Latin America: Opportunities for PARP-Like Inhibitors and Immunotherapy. Genes (Basel) 2019; 10:E786. [PMID: 31658756 PMCID: PMC6827033 DOI: 10.3390/genes10100786] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/28/2019] [Accepted: 10/04/2019] [Indexed: 12/24/2022] Open
Abstract
Germline mutations in BRCA1 and BRCA2 (BRCA1/2) genes are present in about 50% of cases of hereditary breast cancer. Proteins encoded by these genes are key players in DNA repair by homologous recombination (HR). Advances in next generation sequencing and gene panels for breast cancer testing have generated a large amount of data on gene variants implicated in hereditary breast cancer, particularly in genes such as PALB2, ATM, CHEK2, RAD51, MSH2, and BARD1. These genes are involved in DNA repair. Most of these variants have been reported for Caucasian, Jewish, and Asian population, with few reports for other communities, like those in Latin American (LA) countries. We reviewed 81 studies from 11 LA countries published between 2000 and 2019 but most of these studies focused on BRCA1/2 genes. In addition to these genes, breast cancer-related variants have been reported for PALB2, ATM, CHEK2, BARD1, MLH1, BRIP1, MSH2, NBN, MSH6, and PMS2 genes. Some of these variants are unique to LA populations. This analysis may contribute to enhance breast cancer variant characterization, and thus to find therapies and implement precision medicine for LA communities.
Collapse
Affiliation(s)
- Laura Keren Urbina-Jara
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64710, Mexico.
| | - Augusto Rojas-Martinez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64710, Mexico.
| | | | - Dione Aguilar
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64710, Mexico.
- Tecnologico de Monterrey, Centro de Cancer de Mama, Hospital Zambrano Hellion, San Pedro Garza Garcia 66278, Mexico.
| | - Cynthia Villarreal-Garza
- Tecnologico de Monterrey, Centro de Cancer de Mama, Hospital Zambrano Hellion, San Pedro Garza Garcia 66278, Mexico.
- Instituto Nacional de Cancerologia, Departamento de Investigacion, Av. San Fernando #22, Tlalpan, Ciudad de Mexico 14080, Mexico.
| | - Rocio Ortiz-Lopez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64710, Mexico.
| |
Collapse
|
145
|
Abstract
Squamous cell carcinomas (SCCs) arising from aerodigestive or anogenital epithelium that are associated with the human papillomavirus (HPV) are far more readily cured with radiation therapy than HPV-negative SCCs. The mechanism behind this increased radiosensitivity has been proposed to be secondary to defects in DNA repair, although the specific repair pathways that are disrupted have not been elucidated. To gain insight into this important biomarker of radiosensitivity, we first examined genomic patterns reflective of defects in DNA double-strand break repair, comparing HPV-associated and HPV-negative head and neck cancers (HNSCC). Compared to HPV-negative HNSCC genomes, HPV+ cases demonstrated a marked increase in the proportion of deletions with flanking microhomology, a signature associated with a backup, error-prone double-strand break repair pathway known as microhomology-mediated end-joining (MMEJ). Then, using 3 different methodologies to comprehensively profile double-strand break repair pathways in isogenic paired cell lines, we demonstrate that the HPV16 E7 oncoprotein suppresses canonical nonhomologous end-joining (NHEJ) and promotes error-prone MMEJ, providing a mechanistic rationale for the clinical radiosensitivity of these cancers.
Collapse
|
146
|
Feng W, Simpson DA, Carvajal-Garcia J, Price BA, Kumar RJ, Mose LE, Wood RD, Rashid N, Purvis JE, Parker JS, Ramsden DA, Gupta GP. Genetic determinants of cellular addiction to DNA polymerase theta. Nat Commun 2019; 10:4286. [PMID: 31537809 PMCID: PMC6753077 DOI: 10.1038/s41467-019-12234-1] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 08/22/2019] [Indexed: 02/07/2023] Open
Abstract
Polymerase theta (Pol θ, gene name Polq) is a widely conserved DNA polymerase that mediates a microhomology-mediated, error-prone, double strand break (DSB) repair pathway, referred to as Theta Mediated End Joining (TMEJ). Cells with homologous recombination deficiency are reliant on TMEJ for DSB repair. It is unknown whether deficiencies in other components of the DNA damage response (DDR) also result in Pol θ addiction. Here we use a CRISPR genetic screen to uncover 140 Polq synthetic lethal (PolqSL) genes, the majority of which were previously unknown. Functional analyses indicate that Pol θ/TMEJ addiction is associated with increased levels of replication-associated DSBs, regardless of the initial source of damage. We further demonstrate that approximately 30% of TCGA breast cancers have genetic alterations in PolqSL genes and exhibit genomic scars of Pol θ/TMEJ hyperactivity, thereby substantially expanding the subset of human cancers for which Pol θ inhibition represents a promising therapeutic strategy.
Collapse
Affiliation(s)
- Wanjuan Feng
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Dennis A Simpson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Juan Carvajal-Garcia
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Brandon A Price
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Rashmi J Kumar
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Lisle E Mose
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Richard D Wood
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX, 78957, USA
| | - Naim Rashid
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jeremy E Purvis
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Dale A Ramsden
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Gaorav P Gupta
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
147
|
Smith ES, Da Cruz Paula A, Cadoo KA, Abu-Rustum NR, Pei X, Brown DN, Ferrando L, Sebastiao APM, Riaz N, Robson ME, Soslow RA, Reis-Filho JS, Mandelker D, Weigelt B. Endometrial Cancers in BRCA1 or BRCA2 Germline Mutation Carriers: Assessment of Homologous Recombination DNA Repair Defects. JCO Precis Oncol 2019; 3:PO.19.00103. [PMID: 32914019 PMCID: PMC7446423 DOI: 10.1200/po.19.00103] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2019] [Indexed: 01/20/2023] Open
Abstract
PURPOSE Endometrial cancer (EC) is not considered a component of the hereditary breast and ovarian cancer syndrome but can arise in patients with germline BRCA1/2 (gBRCA1/2) mutations. Biallelic BRCA1/2 alterations are associated with genomic features of homologous recombination DNA repair deficiency (HRD) in cancer. We sought to determine if ECs in gBRCA1/2 mutation carriers harbor biallelic alterations and/or features of HRD. METHODS Of 769 patients with EC who underwent germline panel testing, 10 pathogenic gBRCA1/2 mutation carriers were identified, and their tumor- and normal-derived DNA was subjected to massively parallel sequencing targeting at least 410 cancer-related genes. Three gBRCA1/2-associated ECs were identified in 232 ECs subjected to whole-exome sequencing by The Cancer Genome Atlas. Somatic mutations, copy number alterations, loss of heterozygosity, microsatellite instability (MSI), and genomic HRD features were assessed. RESULTS Of the 13 patients included who had EC, eight harbored pathogenic gBRCA1 mutations and five harbored gBRCA2 mutations. Eight (100%) and two (40%) ECs harbored biallelic BRCA1 and BRCA2 alterations through loss of heterozygosity of the wild-type allele. All ECs harbored somatic TP53 mutations. One monoallelic/sporadic gBRCA2-associated EC had MLH1 promoter methylation and was MSI high. High large-scale state transition scores, a genomic feature of HRD, were found only in ECs with bi- but not monoallelic BRCA1/2 alterations. The Signature Multivariate Analysis HRD signature Sig3 was enriched in biallelic gBRCA1/2 ECs, and the three ECs from The Cancer Genome Atlas with BRCA1 biallelic alterations subjected to whole-exome sequencing displayed a dominant HRD-related mutational signature 3. CONCLUSION A subset of gBRCA1/2-associated ECs harbor biallelic BRCA1/2 alterations and genomic features of HRD, which may benefit from homologous recombination-directed treatment regimens. ECs in BRCA2 mutation carriers might be sporadic and even MSI high, and may potentially benefit from immune-checkpoint inhibition.
Collapse
Affiliation(s)
- Evan S. Smith
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | - Xin Pei
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Lorenzo Ferrando
- Memorial Sloan Kettering Cancer Center, New York, NY
- University of Genoa, Genoa, Italy
| | | | - Nadeem Riaz
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | | | | |
Collapse
|
148
|
Jinjia C, Xiaoyu W, Hui S, Wenhua L, Zhe Z, Xiaodong Z, Midie X. The use of DNA repair genes as prognostic indicators of gastric cancer. J Cancer 2019; 10:4866-4875. [PMID: 31598158 PMCID: PMC6775511 DOI: 10.7150/jca.31062] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 07/20/2019] [Indexed: 01/03/2023] Open
Abstract
DNA repair genes can be used as prognostic biomarkers in many types of cancer. We aimed to identify prognostic DNA repair genes in patients with gastric cancer (GC) by systematically bioinformatic approaches using web-based database. Global gene expression profiles from altogether 1,325 GC patients' samples from six independent datasets were included in the study. Clustering analysis was performed to screen potentially abnormal DNA repair genes related to the prognosis of GC, followed by unsupervised clustering analysis to identify molecular subtypes of GC. Characteristics and prognosis differences were analyzed among these molecular subtypes, and modular key genes in molecular subtypes were identified based on changes in expression correlation. Multivariate Cox proportional hazard analysis was used to find the independent prognostic gene. Kaplan-Meier method and log-rank test was used to estimate correlations of key DNA repair genes with GC patients'overall survival. There were 57 key genes significantly associated to GC patients' prognosis, and patients were stratified into three molecular clusters based on their expression profiles, in which patients in Cluster 3 showed the best survival (P < 0.05). After a three-phase training, test and validation process, the expression profile of 13 independent key DNA repair genes were identified can classify the prognostic risk of patients. Compared with patients with low-risk score, patients with high risk score in the training set had shorter overall survival (P < 0.0001). Furthermore, we verified equivalent findings by these key DNA repair genes in the test set (P < 0.0001) and the independent validation set (P = 0.0024). Our results suggest a great potential for the use of DNA repair gene profiling as a powerful marker in prognostication and inform treatment decisions for GC patients.
Collapse
Affiliation(s)
- Chang Jinjia
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Wang Xiaoyu
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Sun Hui
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Li Wenhua
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Zhang Zhe
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Zhu Xiaodong
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Xu Midie
- Department of Pathology & biobank, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| |
Collapse
|
149
|
Li A, Geyer FC, Blecua P, Lee JY, Selenica P, Brown DN, Pareja F, Lee SSK, Kumar R, Rivera B, Bi R, Piscuoglio S, Wen HY, Lozada JR, Gularte-Mérida R, Cavallone L, Rezoug Z, Nguyen-Dumont T, Peterlongo P, Tondini C, Terkelsen T, Rønlund K, Boonen SE, Mannerma A, Winqvist R, Janatova M, Rajadurai P, Xia B, Norton L, Robson ME, Ng PS, Looi LM, Southey MC, Weigelt B, Soo-Hwang T, Tischkowitz M, Foulkes WD, Reis-Filho JS. Homologous recombination DNA repair defects in PALB2-associated breast cancers. NPJ Breast Cancer 2019; 5:23. [PMID: 31428676 PMCID: PMC6687719 DOI: 10.1038/s41523-019-0115-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 06/04/2019] [Indexed: 01/02/2023] Open
Abstract
Mono-allelic germline pathogenic variants in the Partner And Localizer of BRCA2 (PALB2) gene predispose to a high-risk of breast cancer development, consistent with the role of PALB2 in homologous recombination (HR) DNA repair. Here, we sought to define the repertoire of somatic genetic alterations in PALB2-associated breast cancers (BCs), and whether PALB2-associated BCs display bi-allelic inactivation of PALB2 and/or genomic features of HR-deficiency (HRD). Twenty-four breast cancer patients with pathogenic PALB2 germline mutations were analyzed by whole-exome sequencing (WES, n = 16) or targeted capture massively parallel sequencing (410 cancer genes, n = 8). Somatic genetic alterations, loss of heterozygosity (LOH) of the PALB2 wild-type allele, large-scale state transitions (LSTs) and mutational signatures were defined. PALB2-associated BCs were found to be heterogeneous at the genetic level, with PIK3CA (29%), PALB2 (21%), TP53 (21%), and NOTCH3 (17%) being the genes most frequently affected by somatic mutations. Bi-allelic PALB2 inactivation was found in 16 of the 24 cases (67%), either through LOH (n = 11) or second somatic mutations (n = 5) of the wild-type allele. High LST scores were found in all 12 PALB2-associated BCs with bi-allelic PALB2 inactivation sequenced by WES, of which eight displayed the HRD-related mutational signature 3. In addition, bi-allelic inactivation of PALB2 was significantly associated with high LST scores. Our findings suggest that the identification of bi-allelic PALB2 inactivation in PALB2-associated BCs is required for the personalization of HR-directed therapies, such as platinum salts and/or PARP inhibitors, as the vast majority of PALB2-associated BCs without PALB2 bi-allelic inactivation lack genomic features of HRD.
Collapse
Affiliation(s)
- Anqi Li
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
- Department of Pathology, Fudan University Shanghai Cancer Center and Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Felipe C. Geyer
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Pedro Blecua
- Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Ju Youn Lee
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - David N. Brown
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Fresia Pareja
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Simon S. K. Lee
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Rahul Kumar
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Barbara Rivera
- Departments of Oncology and Human Genetics, McGill University, Montreal, Quebec Canada
- Cancer Axis, Lady Davis Institute, Jewish General Hospital, Montreal, Quebec Canada
| | - Rui Bi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
- Department of Pathology, Fudan University Shanghai Cancer Center and Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Salvatore Piscuoglio
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Hannah Y. Wen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - John R. Lozada
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | | | - Luca Cavallone
- Departments of Oncology and Human Genetics, McGill University, Montreal, Quebec Canada
- Cancer Axis, Lady Davis Institute, Jewish General Hospital, Montreal, Quebec Canada
| | - Zoulikha Rezoug
- Cancer Prevention Center, Jewish General Hospital, Montreal, Quebec Canada
| | - Tu Nguyen-Dumont
- Genetic Epidemiology Laboratory, Department of Clinical Pathology, University of Melbourne, Parkville, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Victoria, Australia
| | - Paolo Peterlongo
- IFOM, The Italian Foundation for Cancer Research Institute of Molecular Oncology, Milan, Italy
| | | | - Thorkild Terkelsen
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| | - Karina Rønlund
- Department of Clinical Genetics, Vejle Hospital, Vejle, Denmark
| | - Susanne E. Boonen
- Clinical Genetics Unit, Department of Pediatrics, Zealand University Hospital, Roskilde, Denmark
| | - Arto Mannerma
- Biocenter Kuopio and Cancer Center of Easter Finland, University of Eastern Finland, Kuopio, Finland
| | - Robert Winqvist
- Laboratory of Cancer Genetics and Tumor Biology, Cancer and Translational Medicine Research Unit, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Marketa Janatova
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Bing Xia
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ USA
| | - Larry Norton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Mark E. Robson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Pei-Sze Ng
- Cancer Research Malaysia, Subang Jaya, Malaysia
| | - Lai-Meng Looi
- Department of Pathology, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - Melissa C. Southey
- Genetic Epidemiology Laboratory, Department of Clinical Pathology, University of Melbourne, Parkville, Victoria, Australia
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Teo Soo-Hwang
- Cancer Research Malaysia, Subang Jaya, Malaysia
- University Malaya Cancer Research Institute, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - Marc Tischkowitz
- Department of Medical Genetics, University of Cambridge, Cambridge, UK
| | - William D. Foulkes
- Cancer Axis, Lady Davis Institute, Jewish General Hospital, Montreal, Quebec Canada
- Cancer Prevention Center, Jewish General Hospital, Montreal, Quebec Canada
- Cancer Program, Research Institute McGill University Health Centre, Montreal, Quebec Canada
| | - Jorge S. Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| |
Collapse
|
150
|
Hoppe MM, Sundar R, Tan DSP, Jeyasekharan AD. Biomarkers for Homologous Recombination Deficiency in Cancer. J Natl Cancer Inst 2019; 110:704-713. [PMID: 29788099 DOI: 10.1093/jnci/djy085] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/06/2018] [Indexed: 12/11/2022] Open
Abstract
Defective DNA repair is a common hallmark of cancer. Homologous recombination is a DNA repair pathway of clinical interest due to the sensitivity of homologous recombination-deficient cells to poly-ADP ribose polymerase (PARP) inhibitors. The measurement of homologous recombination deficiency (HRD) in cancer is therefore vital to the appropriate design of clinical trials incorporating PARP inhibitors. However, methods to identify HRD in tumors are varied and controversial. Understanding existing and new methods to measure HRD is important to their appropriate use in clinical trials and practice. The aim of this review is to summarize the biology and clinical validation of current methods to measure HRD, to aid decision-making for patient stratification and translational research in PARP inhibitor trials. We discuss the current clinical development of PARP inhibitors, along with established indicators for HRD such as germline BRCA1/2 mutation status and clinical response to platinum-based therapy. We then examine newer assays undergoing clinical validation, including 1) somatic mutations in homologous recombination genes, 2) "genomic scar" assays using array-based comparative genomic hybridization (aCGH), single nucleotide polymorphism (SNP) analysis or mutational signatures derived from next-generation sequencing, 3) transcriptional profiles of HRD, and 4) phenotypic or functional assays of protein expression and localization. We highlight the strengths and weaknesses of each of these assays, for consideration during the design of studies involving PARP inhibitors.
Collapse
Affiliation(s)
- Michal M Hoppe
- Cancer Science Institute of Singapore, National University Hospital, Singapore
| | - Raghav Sundar
- Department of Haematology-Oncology, National University Hospital, Singapore
| | - David S P Tan
- Cancer Science Institute of Singapore, National University Hospital, Singapore.,Department of Haematology-Oncology, National University Hospital, Singapore
| | - Anand D Jeyasekharan
- Cancer Science Institute of Singapore, National University Hospital, Singapore.,Department of Haematology-Oncology, National University Hospital, Singapore
| |
Collapse
|