1
|
Saponaro C, Damato M, Stanca E, Aboulouard S, Zito FA, De Summa S, Traversa D, Schirosi L, Bravaccini S, Pirini F, Fonzi E, Tebaldi M, Puccetti M, Gaballo A, Pantalone L, Ronci M, Magnani L, Sergi D, Tinelli A, Tacconi S, Siculella L, Giudetti AM, Fournier I, Salzet M, Trerotola M, Vergara D. Unraveling the protein kinase C/NDRG1 signaling network in breast cancer. Cell Biosci 2024; 14:156. [PMID: 39736699 DOI: 10.1186/s13578-024-01336-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/09/2024] [Indexed: 01/01/2025] Open
Abstract
N-myc downstream-regulated gene 1 (NDRG1) is a member of the NDRG family of intracellular proteins and plays a central role in a wide range of biological processes including stress response, differentiation, and metabolism. The overexpression of NDRG1 is an indicator of poor prognosis in various types of cancer. Here, we found that NDRG1 is an independent prognostic marker of poor outcome in breast cancer (BC). Analysis of the TCGA dataset showed a significant positive correlation between NDRG1 and PRKCA expression, suggesting a mechanistic role of protein kinase C (PKC) in the regulation of NDRG1. We then assessed the hypothesis that PKC might modulate the activity of NDRG1, and observed that different acute stress conditions converging on PKC activation lead to enhanced NDRG1 expression. This mechanism was found to be specific for NDRG1 as the expression of other NDRG members was not affected. Moreover, CRISPR-based inhibition of NDRG1 expression was obtained in a BC cell line, and showed that this protein is a key driver of BC cell invasion through the Rho-associated coiled-coil containing protein kinase 1 (ROCK1)/phosphorylated cofilin pathway that regulates stress fiber assembly, and the modulation of extracellular matrix reorganization related genes. Together, our findings highlight the potential of NDRG1 as a new BC biomarker and uncover a novel mechanism of regulation of NDRG1 expression that might lead to innovative therapeutic strategies.
Collapse
Affiliation(s)
- C Saponaro
- Pathology Department, IRCCS Istituto Tumori "Giovanni Paolo II", 70124, Bari, Italy
| | - M Damato
- Department of Experimental Medicine, University of Salento, Lecce, Italy
| | - E Stanca
- Department of Experimental Medicine, University of Salento, Lecce, Italy
| | - S Aboulouard
- Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille University, Inserm, CHU Lille, U1192, Lille, France
| | - F A Zito
- Pathology Department, IRCCS Istituto Tumori "Giovanni Paolo II", 70124, Bari, Italy
| | - S De Summa
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori "Giovanni Paolo II", 70124, Bari, Italy
| | - D Traversa
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori "Giovanni Paolo II", 70124, Bari, Italy
| | - L Schirosi
- Pathology Department, IRCCS Istituto Tumori "Giovanni Paolo II", 70124, Bari, Italy
| | - S Bravaccini
- Department of Medicine and Surgery, University of Enna "Kore", 94100, Enna, Italy
| | - F Pirini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - E Fonzi
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - M Tebaldi
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - M Puccetti
- Azienda Unità Sanitaria Locale di Imola, Imola, Italy
| | - A Gaballo
- CNR Nanotec, Institute of Nanotechnology, Via Monteroni, 73100, Lecce, Italy
| | - L Pantalone
- Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - M Ronci
- Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - L Magnani
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- Department of Surgery and Cancer, Imperial College London, London, UK
- Department of Oncology and Haemato-Oncology, Università Degli Studi di Milano, Milan, Italy
| | - D Sergi
- Department of Radiology, V. Fazzi Hospital, 73100, Lecce, Italy
| | - A Tinelli
- Department of Obstetrics and Gynecology and CERICSAL, (CEntro di RIcerca Clinico SALentino), "Veris Delli Ponti Hospital", 73020, ScorranoScorrano (Lecce), Italy
| | - S Tacconi
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, P.Le Aldo Moro 5, 00185, Rome, Italy
| | - L Siculella
- Department of Experimental Medicine, University of Salento, Lecce, Italy
| | - A M Giudetti
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| | - I Fournier
- Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille University, Inserm, CHU Lille, U1192, Lille, France
| | - M Salzet
- Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille University, Inserm, CHU Lille, U1192, Lille, France
| | - M Trerotola
- Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.
| | - D Vergara
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy.
| |
Collapse
|
2
|
Sellars E, Savguira M, Wu J, Cancelliere S, Jen M, Krishnan R, Hakem A, Barsyte-Lovejoy D, Hakem R, Narod SA, Kotsopoulos J, Salmena L. A high-throughput approach to identify BRCA1-downregulating compounds to enhance PARP inhibitor sensitivity. iScience 2024; 27:110180. [PMID: 38993666 PMCID: PMC11238136 DOI: 10.1016/j.isci.2024.110180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/29/2024] [Accepted: 06/01/2024] [Indexed: 07/13/2024] Open
Abstract
PARP inhibitors (PARPi) are efficacious in BRCA1-null tumors; however, their utility is limited in tumors with functional BRCA1. We hypothesized that pharmacologically reducing BRCA1 protein levels could enhance PARPi effectiveness in BRCA1 wild-type tumors. To identify BRCA1 downregulating agents, we generated reporter cell lines using CRISPR-mediated editing to tag endogenous BRCA1 protein with HiBiT. These reporter lines enable the sensitive measurement of BRCA1 protein levels by luminescence. Validated reporter cells were used in a pilot screen of epigenetic-modifying probes and a larger screen of more than 6,000 compounds. We identified 7 compounds that could downregulate BRCA1-HiBiT expression and synergize with olaparib. Three compounds, N-acetyl-N-acetoxy chlorobenzenesulfonamide (NANAC), A-443654, and CHIR-124, were validated to reduce BRCA1 protein levels and sensitize breast cancer cells to the toxic effects of olaparib. These results suggest that BRCA1-HiBiT reporter cells hold promise in developing agents to improve the clinical utility of PARPi.
Collapse
Affiliation(s)
- Erin Sellars
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Women's College Research Institute, Women's College Hospital, Toronto, ON M5S 1B2, Canada
| | - Margarita Savguira
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jie Wu
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sabrina Cancelliere
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Mark Jen
- Lunenfeld-Tanenbaum Research Institute, Network Biology Collaborative Centre, High-Throughput Screening, Mt. Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Rehna Krishnan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Anne Hakem
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Dalia Barsyte-Lovejoy
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Structural Genomics Consortium, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Razqallah Hakem
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Steven A Narod
- Women's College Research Institute, Women's College Hospital, Toronto, ON M5S 1B2, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Joanne Kotsopoulos
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Women's College Research Institute, Women's College Hospital, Toronto, ON M5S 1B2, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Leonardo Salmena
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Women's College Research Institute, Women's College Hospital, Toronto, ON M5S 1B2, Canada
| |
Collapse
|
3
|
Huang Y, Ren S, Ding L, Jiang Y, Luo J, Huang J, Yin X, Zhao J, Fu S, Liao J. TP53-specific mutations serve as a potential biomarker for homologous recombination deficiency in breast cancer: a clinical next-generation sequencing study. PRECISION CLINICAL MEDICINE 2024; 7:pbae009. [PMID: 38745917 PMCID: PMC11092399 DOI: 10.1093/pcmedi/pbae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/07/2024] [Indexed: 05/16/2024] Open
Abstract
Background TP53 mutations and homologous recombination deficiency (HRD) occur frequently in breast cancer. However, the characteristics of TP53 pathogenic mutations in breast cancer patients with/without HRD are not clear. Methods Clinical next-generation sequencing (NGS) of both tumor and paired blood DNA from 119 breast cancer patients (BRCA-119 cohort) was performed with a 520-gene panel. Mutations, tumor mutation burden (TMB), and genomic HRD scores were assessed from NGS data. NGS data from 47 breast cancer patients in the HRD test cohort were analyzed for further verification. Results All TP53 pathogenic mutations in patients had somatic origin, which was associated with the protein expression of estrogen receptor and progestogen receptor. Compared to patients without TP53 pathologic mutations, patients with TP53 pathologic mutations had higher levels of HRD scores and different genomic alterations. The frequency of TP53 pathologic mutation was higher in the HRD-high group (HRD score ≥ 42) relative to that in the HRD-low group (HRD score < 42). TP53 has different mutational characteristics between the HRD-low and HRD-high groups. TP53-specific mutation subgroups had diverse genomic features and TMB. Notably, TP53 pathogenic mutations predicted the HRD status of breast cancer patients with an area under the curve (AUC) of 0.61. TP53-specific mutations, namely HRD-low mutation, HRD-high mutation, and HRD common mutation, predicted the HRD status of breast cancer patients with AUC values of 0.32, 0.72, and 0.58, respectively. Interestingly, TP53 HRD-high mutation and HRD common mutation combinations showed the highest AUC values (0.80) in predicting HRD status. Conclusions TP53-specific mutation combinations predict the HRD status of patients, indicating that TP53 pathogenic mutations could serve as a potential biomarker for poly-ADP-ribose polymerase (PARP) inhibitors in breast cancer patients .
Collapse
Affiliation(s)
- Yongsheng Huang
- Cellular & Molecular Diagnostics Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Shuwei Ren
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Linxiaoxiao Ding
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangzhou Regenerative Medicine and Health, Guangdong Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yuanling Jiang
- Cellular & Molecular Diagnostics Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jiahuan Luo
- Cellular & Molecular Diagnostics Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jinghua Huang
- Cellular & Molecular Diagnostics Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xinke Yin
- Cellular & Molecular Diagnostics Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jianli Zhao
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangzhou Regenerative Medicine and Health, Guangdong Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Sha Fu
- Cellular & Molecular Diagnostics Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jianwei Liao
- Cellular & Molecular Diagnostics Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| |
Collapse
|
4
|
Triple negative breast cancer: approved treatment options and their mechanisms of action. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04189-6. [PMID: 35976445 DOI: 10.1007/s00432-022-04189-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/06/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE Breast cancer, the most prevalent cancer worldwide, consists of 4 main subtypes, namely, Luminal A, Luminal B, HER2-positive, and Triple-negative breast cancer (TNBC). Triple-negative breast tumors, which do not express estrogen, progesterone, and HER2 receptors, account for approximately 15-20% of breast cancer cases. The lack of traditional receptor targets contributes to the heterogenous, aggressive, and refractory nature of these tumors, resulting in limited therapeutic strategies. METHODS Chemotherapeutics such as taxanes and anthracyclines have been the traditional go to treatment regimens for TNBC patients. Paclitaxel, docetaxel, doxorubicin, and epirubicin have been longstanding, Food and Drug Administration (FDA)-approved therapies against TNBC. Additionally, the FDA approved PARP inhibitors such as olaparib and atezolizumab to be used in combination with chemotherapies, primarily to improve their efficiency and reduce adverse patient outcomes. The immunotherapeutic Keytruda was the latest addition to the FDA-approved list of drugs used to treat TNBC. RESULTS The following review aims to elucidate current FDA-approved therapeutics and their mechanisms of action, shedding a light on the various strategies currently used to circumvent the treatment-resistant nature of TNBC cases. CONCLUSION The recent approval and use of therapies such as Trodelvy, olaparib and Keytruda has its roots in the development of an understanding of signaling pathways that drive tumour growth. In the future, the emergence of novel drug delivery methods may help increase the efficiency of these therapies whiel also reducing adverse side effects.
Collapse
|
5
|
Loss of function of BRCA1 promotes EMT in mammary tumors through activation of TGFβR2 signaling pathway. Cell Death Dis 2022; 13:195. [PMID: 35236825 PMCID: PMC8891277 DOI: 10.1038/s41419-022-04646-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 01/25/2022] [Accepted: 02/09/2022] [Indexed: 12/15/2022]
Abstract
BRCA1 deficient breast cancers are aggressive and chemoresistant due, in part, to their enrichment of cancer stem cells that can be generated from carcinoma cells by an epithelial-mesenchymal transition (EMT). We previously discovered that BRCA1 deficiency activates EMT in mammary tumorigenesis. How BRCA1 controls EMT and how to effectively target BRCA1-deficient cancers remain elusive. We analyzed murine and human tumors and identified a role for Tgfβr2 in governing the molecular aspects of EMT that occur with Brca1 loss. We utilized CRISPR to delete Tgfβr2 and specific inhibitors to block Tgfβr2 activity and followed up with the molecular analysis of assays for tumor growth and metastasis. We discovered that heterozygous germline deletion, or epithelia-specific deletion of Brca1 in mice, activates Tgfβr2 signaling pathways in mammary tumors. BRCA1 depletion promotes TGFβ-mediated EMT activation in cancer cells. BRCA1 binds to the TGFβR2 locus to repress its transcription. Targeted deletion or pharmaceutical inhibition of Tgfβr2 in Brca1-deficient tumor cells reduces EMT and suppresses tumorigenesis and metastasis. BRCA1 and TGFβR2 expression levels are inversely related in human breast cancers. This study reveals for the first time that a targetable TGFβR signaling pathway is directly activated by BRCA1-deficiency in the induction of EMT in breast cancer progression.
Collapse
|
6
|
Shafaee MN, Makawita S, Lim B, Ellis MJ, Ducan DL, Ludwig MS, Duncan DL. Concurrent Chemo-radiation As a Means of Achieving Pathologic Complete Response in Triple Negative Breast Cancer. Clin Breast Cancer 2021; 22:e536-e543. [DOI: 10.1016/j.clbc.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 11/16/2021] [Accepted: 12/03/2021] [Indexed: 11/03/2022]
|
7
|
Bai F, Zhang LH, Liu X, Wang C, Zheng C, Sun J, Li M, Zhu WG, Pei XH. GATA3 functions downstream of BRCA1 to suppress EMT in breast cancer. Theranostics 2021; 11:8218-8233. [PMID: 34373738 PMCID: PMC8344017 DOI: 10.7150/thno.59280] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
Purpose: Functional loss of BRCA1 is associated with poorly differentiated and metastatic breast cancers that are enriched with cancer stem cells (CSCs). CSCs can be generated from carcinoma cells through an epithelial-mesenchymal transition (EMT) program. We and others have previously demonstrated that BRCA1 suppresses EMT and regulates the expression of multiple EMT-related transcription factors. However, the downstream mediators of BRCA1 function in EMT suppression remain elusive. Methods: Depletion of BRCA1 or GATA3 activates p18INK4C , a cell cycle inhibitor which inhibits mammary epithelial cell proliferation. We have therefore created genetically engineered mice with Brca1 or Gata3 loss in addition to deletion of p18INK4C , to rescue proliferative defects caused by deficiency of Brca1 or Gata3. By using these mutant mice along with human BRCA1 deficient as well as proficient breast cancer tissues and cells, we investigated and compared the role of Brca1 and Gata3 loss in the activation of EMT in breast cancers. Results: We discovered that BRCA1 and GATA3 expressions were positively correlated in human breast cancer. Depletion of BRCA1 stimulated methylation of GATA3 promoter thereby repressing GATA3 transcription. We developed Brca1 and Gata3 deficient mouse system. We found that Gata3 deficiency in mice induced poorly-differentiated mammary tumors with the activation of EMT and promoted tumor initiating and metastatic potential. Gata3 deficient mammary tumors phenocopied Brca1 deficient tumors in the induction of EMT under the same genetic background. Reconstitution of Gata3 in Brca1-deficient tumor cells activated mesenchymal-epithelial transition, suppressing tumor initiation and metastasis. Conclusions: Our finding, for the first time, demonstrates that GATA3 functions downstream of BRCA1 to suppress EMT in controlling mammary tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Feng Bai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- Department of Pathology, Shenzhen University Health Science Center, Shenzhen 518060, China
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136, USA
| | - Li-Han Zhang
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136, USA
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China
- The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, China
| | - Xiong Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Chuying Wang
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136, USA
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Chenglong Zheng
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Jianping Sun
- Department of Mathematics and Statistics, University of North Carolina at Greensboro, Greensboro, NC 27402, USA
| | - Min Li
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Xin-Hai Pei
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136, USA
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
| |
Collapse
|
8
|
Wu Y, Pan X, Dou J, Zhang Q, Li Y, Sheng Y, Liu X. A novel germline BRCA1 mutation identified in a family with hereditary breast and ovarian cancer syndrome. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2021; 15:11795549211028569. [PMID: 34276234 PMCID: PMC8255550 DOI: 10.1177/11795549211028569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/08/2021] [Indexed: 12/15/2022]
Abstract
Pathogenic germline mutations occurring in the BRCA1 (MIM:113705) and BRCA2 (MIM: 600185), which always result in truncated protein or nonsense-mediated mRNA decay, have been identified to increase the risk of hereditary breast, ovarian, pancreatic, prostate, and melanoma cancers. Recent studies show that BRCA1/2 germline mutations also contribute to half of all hereditary breast and ovarian cancer (HBOC). In this case series, we reported a novel frameshift mutation of the BRCA1 gene. This novel frameshift mutation occurs in exon10 of BRCA1 and may result in a lack of the serine cluster domain and BRCA1 C-terminus domain, which mediates the function of BRCA1 in DNA repair and are responsible for activation function of BRCA1. The mutation was present in a Chinese hereditary male/female breast and ovarian cancer family characterized by a high incidence of breast cancer and/or ovarian cancer among the relatives and by a high incidence of triple negative breast cancer (TNBC). Our findings speculate that BRCA1 E1148Rfs*7 mutation may be related to the occurrence of HBOC and even TNBC. Interestingly, three cases of TNBC with this novel BRCA1 mutation in this case series showed a good disease-free survival, one of them has a disease-free survival up to 7 years. Therefore, further study is required to confirm that whether this mutation is associated with good prognosis of HBOC.
Collapse
Affiliation(s)
- Yanmei Wu
- Department of Breast and Thyroid Surgery, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Xiaodong Pan
- Dinfectome Medical Technology Inc., Shanghai, China
| | - Juan Dou
- Department of Breast and Thyroid Surgery, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Quan Zhang
- Department of Breast and Thyroid Surgery, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Yuantong Li
- Department of Biology, Brandeis University, Waltham, MA, USA
| | - Yuan Sheng
- Department of Breast and Thyroid Surgery, Changhai Hospital, Navy Medical University, Shanghai, China
- Yuan Sheng and Xishui Liu, Department of Breast and Thyroid Surgery, Changhai Hospital, Navy Medical University, 168 Changhai Road, Shanghai 200433, China. Emails: and
| | - Xishui Liu
- Department of Breast and Thyroid Surgery, Changhai Hospital, Navy Medical University, Shanghai, China
| |
Collapse
|
9
|
Spiegel JO, Van Houten B, Durrant JD. PARP1: Structural insights and pharmacological targets for inhibition. DNA Repair (Amst) 2021; 103:103125. [PMID: 33940558 PMCID: PMC8206044 DOI: 10.1016/j.dnarep.2021.103125] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/24/2021] [Accepted: 04/09/2021] [Indexed: 12/25/2022]
Abstract
Poly(ADP-ribose) polymerase 1 (PARP1, also known as ADPRT1) is a multifunctional human ADP-ribosyltransferase. It plays a role in multiple DNA repair pathways, including the base excision repair (BER), non-homologous end joining (NHEJ), homologous recombination (HR), and Okazaki-fragment processing pathways. In response to DNA strand breaks, PARP1 covalently attaches ADP-ribose moieties to arginine, glutamate, aspartate, cysteine, lysine, and serine acceptor sites on both itself and other proteins. This signal recruits DNA repair proteins to the site of DNA damage. PARP1 binding to these sites enhances ADP-ribosylation via allosteric communication between the distant DNA binding and catalytic domains. In this review, we provide a general overview of PARP1 and emphasize novel potential approaches for pharmacological inhibition. Clinical PARP1 inhibitors bind the catalytic pocket, where they directly interfere with ADP-ribosylation. Some inhibitors may further enhance potency by "trapping" PARP1 on DNA via an allosteric mechanism, though this proposed mode of action remains controversial. PARP1 inhibitors are used clinically to treat some cancers, but resistance is common, so novel pharmacological approaches are urgently needed. One approach may be to design novel small molecules that bind at inter-domain interfaces that are essential for PARP1 allostery. To illustrate these points, this review also includes instructive videos showing PARP1 structures and mechanisms.
Collapse
Affiliation(s)
- Jacob O Spiegel
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Bennett Van Houten
- UPMC-Hillman Cancer Center, Pittsburgh, PA, 15232, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jacob D Durrant
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
10
|
Naik A, Decock J. Targeting of lactate dehydrogenase C dysregulates the cell cycle and sensitizes breast cancer cells to DNA damage response targeted therapy. Mol Oncol 2021; 16:885-903. [PMID: 34050611 PMCID: PMC8847988 DOI: 10.1002/1878-0261.13024] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/21/2021] [Accepted: 05/27/2021] [Indexed: 12/31/2022] Open
Abstract
The cancer testis antigen (CTA) lactate dehydrogenase C (LDHC) is a promising anticancer target with tumor-specific expression and immunogenicity. Interrogation of breast cancer patient cohorts from The Cancer Genome Atlas (TCGA) and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) indicate that upregulation of LDHC expression correlates with unfavorable prognosis. Although the role of LDHC is well characterized in spermatocytes, its role in tumors remains largely unknown. We investigated whether LDHC is involved in regulating genomic stability and whether it could be targeted to affect tumor cellular fitness. Silencing LDHC in four breast cancer cell lines significantly increased the presence of giant cells, nuclear aberrations, DNA damage, and apoptosis. LDHC-silenced cells demonstrated aberrant cell cycle progression with differential expression of cell cycle checkpoint and DNA damage response regulators. In addition, LDHC silencing-induced microtubule destabilization, culminating in increased mitotic catastrophe and reduced long-term survival. Notably, the clonogenicity of LDHC-silenced cells was further reduced by treatment with the poly (ADP-ribose) polymerase (PARP) inhibitor olaparib and with the DNA-damaging drug cisplatin. This study supports the therapeutic potential of targeting LDHC to mitigate cancer cell survival and improve sensitivity to agents that cause DNA damage or inhibit its repair.
Collapse
Affiliation(s)
- Adviti Naik
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Julie Decock
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
11
|
Chen Z, Wang X, Li X, Zhou Y, Chen K. Deep exploration of PARP inhibitors in breast cancer: monotherapy and combination therapy. J Int Med Res 2021; 49:300060521991019. [PMID: 33541181 PMCID: PMC8164563 DOI: 10.1177/0300060521991019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 01/05/2021] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Nearly 5% of patients with breast cancer carry germline BRCA mutations, which are more common in triple-negative breast cancer (TNBC). Previous clinical trials demonstrated the therapeutic efficacy of poly (ADP-ribose) polymerase inhibitors (PARPis) against BRCA-mutated metastatic breast cancer. The current study conducted a systemic review and meta-analysis of the clinical efficiency and safety of PARPis, either alone or combined with chemotherapy, in patients with TNBC. METHODS We searched PubMed, EMBASE, and ClinicalTrials.gov to identify randomized controlled trials comparing PARPi therapy with chemotherapy, and comparisons of chemotherapy plus PARPis with chemotherapy alone were included. The study endpoints included the clinical response, progression-free survival, and adverse event rates. RESULTS PARPi therapy was revealed to improve progression-free survival in patients with advanced breast cancer, either alone or in combination with chemotherapy. Subgroup analysis illustrated that patients with mutant BRCA1 and mutant BRCA2 and those who had not been treated with platinum-based agents could specifically benefit from PARPis. CONCLUSION PARPi monotherapy can significantly improve clinical outcomes in patients with advanced breast cancer, especially those with TNBC, those who had not previously received platinum therapy, and those with mutant BRCA1/2. PARPis combined with chemotherapy represent new treatment options for patients with advanced cancer.
Collapse
Affiliation(s)
- Zheling Chen
- Department of Medical Oncology, Zhejiang Provincial People’s
Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang,
People’s Republic of China
| | - Xiao Wang
- Department of Medical Oncology, Zhejiang Provincial People’s
Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang,
People’s Republic of China
| | - Xiao Li
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang
Provincial People's Hospital, People's Hospital of Hangzhou Medical College,
Hangzhou, Zhejiang, People’s Republic of China
| | - Yucheng Zhou
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang
Provincial People's Hospital, People's Hospital of Hangzhou Medical College,
Hangzhou, Zhejiang, People’s Republic of China
| | - Ke Chen
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang
Provincial People's Hospital, People's Hospital of Hangzhou Medical College,
Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
12
|
Bai F, Liu S, Liu X, Hollern DP, Scott A, Wang C, Zhang L, Fan C, Fu L, Perou CM, Zhu WG, Pei XH. PDGFRβ is an essential therapeutic target for BRCA1-deficient mammary tumors. Breast Cancer Res 2021; 23:10. [PMID: 33478572 PMCID: PMC7819225 DOI: 10.1186/s13058-021-01387-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/03/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Basal-like breast cancers (BLBCs) are a leading cause of cancer death due to their capacity to metastasize and lack of effective therapies. More than half of BLBCs have a dysfunctional BRCA1. Although most BRCA1-deficient cancers respond to DNA-damaging agents, resistance and tumor recurrence remain a challenge to survival outcomes for BLBC patients. Additional therapies targeting the pathways aberrantly activated by BRCA1 deficiency are urgently needed. METHODS Most BRCA1-deficient BLBCs carry a dysfunctional INK4-RB pathway. Thus, we created genetically engineered mice with Brca1 loss and deletion of p16INK4A, or separately p18INK4C, to model the deficient INK4-RB signaling in human BLBC. By using these mutant mice and human BRCA1-deficient and proficient breast cancer tissues and cells, we tested if there exists a druggable target in BRCA1-deficient breast cancers. RESULTS Heterozygous germline or epithelium-specific deletion of Brca1 in p18INK4C- or p16INK4A-deficient mice activated Pdgfrβ signaling, induced epithelial-to-mesenchymal transition, and led to BLBCs. Confirming this role, targeted deletion of Pdgfrβ in Brca1-deficient tumor cells promoted cell death, induced mesenchymal-to-epithelial transition, and suppressed tumorigenesis. Importantly, we also found that pharmaceutical inhibition of Pdgfrβ and its downstream target Pkcα suppressed Brca1-deficient tumor initiation and progression and effectively killed BRCA1-deficient cancer cells. CONCLUSIONS Our work offers the first genetic and biochemical evidence that PDGFRβ-PKCα signaling is repressed by BRCA1, which establishes PDGFRβ-PKCα signaling as a therapeutic target for BRCA1-deficient breast cancers.
Collapse
Affiliation(s)
- Feng Bai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China
- Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Shiqin Liu
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xiong Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Daniel P Hollern
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alexandria Scott
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Chuying Wang
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lihan Zhang
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
- The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Cheng Fan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Li Fu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China
- Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, 518039, China
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Xin-Hai Pei
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China.
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA.
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, 518060, China.
| |
Collapse
|
13
|
Wang H, Ren B, Liu Y, Jiang B, Guo Y, Wei M, Luo L, Kuang X, Qiu M, Lv L, Xu H, Qi R, Yan H, Xu D, Wang Z, Huo CX, Zhu Y, Zhao Y, Wu Y, Qin Z, Su D, Tang T, Wang F, Sun X, Feng Y, Peng H, Wang X, Gao Y, Liu Y, Gong W, Yu F, Liu X, Wang L, Zhou C. Discovery of Pamiparib (BGB-290), a Potent and Selective Poly (ADP-ribose) Polymerase (PARP) Inhibitor in Clinical Development. J Med Chem 2020; 63:15541-15563. [DOI: 10.1021/acs.jmedchem.0c01346] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
14
|
Wang Y, Park JYP, Pacis A, Denroche RE, Jang GH, Zhang A, Cuggia A, Domecq C, Monlong J, Raitses-Gurevich M, Grant RC, Borgida A, Holter S, Stossel C, Bu S, Masoomian M, Lungu IM, Bartlett JM, Wilson JM, Gao ZH, Riazalhosseini Y, Asselah J, Bouganim N, Cabrera T, Boucher LM, Valenti D, Biagi J, Greenwood CM, Polak P, Foulkes WD, Golan T, O'Kane GM, Fischer SE, Knox JJ, Gallinger S, Zogopoulos G. A Preclinical Trial and Molecularly Annotated Patient Cohort Identify Predictive Biomarkers in Homologous Recombination–deficient Pancreatic Cancer. Clin Cancer Res 2020; 26:5462-5476. [DOI: 10.1158/1078-0432.ccr-20-1439] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/24/2020] [Accepted: 08/03/2020] [Indexed: 12/27/2022]
Abstract
Abstract
Purpose:
Pancreatic ductal adenocarcinoma (PDAC) arising in patients with a germline BRCA1 or BRCA2 (gBRCA) mutation may be sensitive to platinum and PARP inhibitors (PARPi). However, treatment stratification based on gBRCA mutational status alone is associated with heterogeneous responses.
Experimental Design:
We performed a seven-arm preclinical trial consisting of 471 mice, representing 12 unique PDAC patient-derived xenografts, of which nine were gBRCA mutated. From 179 patients whose PDAC was whole-genome and transcriptome sequenced, we identified 21 cases with homologous recombination deficiency (HRD), and investigated prognostic biomarkers.
Results:
We found that biallelic inactivation of BRCA1/BRCA2 is associated with genomic hallmarks of HRD and required for cisplatin and talazoparib (PARPi) sensitivity. However, HRD genomic hallmarks persisted in xenografts despite the emergence of therapy resistance, indicating the presence of a genomic scar. We identified tumor polyploidy and a low Ki67 index as predictors of poor cisplatin and talazoparib response. In patients with HRD PDAC, tumor polyploidy and a basal-like transcriptomic subtype were independent predictors of shorter survival. To facilitate clinical assignment of transcriptomic subtype, we developed a novel pragmatic two-marker assay (GATA6:KRT17).
Conclusions:
In summary, we propose a predictive and prognostic model of gBRCA-mutated PDAC on the basis of HRD genomic hallmarks, Ki67 index, tumor ploidy, and transcriptomic subtype.
Collapse
Affiliation(s)
- Yifan Wang
- 1Rosalind and Morris Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
- 2Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jin Yong Patrick Park
- 1Rosalind and Morris Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
- 2Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Alain Pacis
- 1Rosalind and Morris Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
- 3Canadian Centre for Computational Genomics, McGill University and Genome Quebec Innovation Center, Montreal, Quebec, Canada
| | | | - Gun Ho Jang
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Amy Zhang
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Adeline Cuggia
- 1Rosalind and Morris Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
- 2Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Celine Domecq
- 1Rosalind and Morris Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
- 2Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jean Monlong
- 5Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Maria Raitses-Gurevich
- 6Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Robert C. Grant
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- 7Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Ayelet Borgida
- 8Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Spring Holter
- 8Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Chani Stossel
- 6Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- 9Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Simeng Bu
- 1Rosalind and Morris Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
- 2Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Mehdi Masoomian
- 10Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Ilinca M. Lungu
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - John M.S. Bartlett
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- 10Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Julie M. Wilson
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Zu-Hua Gao
- 11Department of Pathology, McGill University, Montreal, Quebec, Canada
| | | | - Jamil Asselah
- 12Department of Oncology, McGill University, Montreal, Quebec, Canada
| | | | - Tatiana Cabrera
- 13Department of Diagnostic Radiology, McGill University, Montreal, Quebec, Canada
| | - Louis-Martin Boucher
- 13Department of Diagnostic Radiology, McGill University, Montreal, Quebec, Canada
| | - David Valenti
- 13Department of Diagnostic Radiology, McGill University, Montreal, Quebec, Canada
| | - James Biagi
- 14Department of Oncology, Queen's University, Kingston, Ontario, Canada
| | - Celia M.T. Greenwood
- 5Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- 12Department of Oncology, McGill University, Montreal, Quebec, Canada
- 15Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
- 16Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Paz Polak
- 17Icahn School of Medicine at Mount Sinai Hospital, New York, New York
| | - William D. Foulkes
- 2Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- 5Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Talia Golan
- 6Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- 9Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Grainne M. O'Kane
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- 7Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Sandra E. Fischer
- 10Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer J. Knox
- 7Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Steven Gallinger
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- 7Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - George Zogopoulos
- 1Rosalind and Morris Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
- 2Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
15
|
Shaikh SS, Emens LA. Current and emerging biologic therapies for triple negative breast cancer. Expert Opin Biol Ther 2020; 22:591-602. [PMID: 32713217 DOI: 10.1080/14712598.2020.1801627] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Triple negative breast cancer, defined by a lack of estrogen receptor, progesterone receptor, or human epidermal growth factor2, accounts for approximately 15% of breast cancer patients. Treatment options have historically been limited to chemotherapy, which has significant toxicity and a suboptimal impact on the five-year relapse rate and survival. AREAS COVERED Transcriptomic analyses reveal that TNBC is biologically heterogenous. Predictive biomarkers based on the distinct biology of the different subtypes of TNBC should identify patients that will derive the greatest benefit from a specifically targeted therapeutic agent. Two biomarker-driven treatments have recently been approved: poly-ADP ribose polymerase inhibitors for patients with germline BRCA mutations and atezolizumab in combination with nab-paclitaxel for patients expressing PD-L1 on tumor-infiltrating immune cells. EXPERT OPINION Identifying informative predictive biomarkers is critical for the optimal development of targeted drugs for TNBC. Some targeted agents, such as the antibody-drug conjugate sacituzumab govitecan-hziy and the precision medicines capivasertib and ipatisertib, have already shown promising results in early clinical trials, and the results of definitive phase 3 trials are eagerly awaited. Additionally, testing novel immunotherapies and other targeted agents in earlier stages of disease, particularly the neoadjuvant setting, is a high priority.
Collapse
Affiliation(s)
- Saba S Shaikh
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Leisha A Emens
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh, Pittsburgh, PA, USA.,UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
16
|
Choi C, Cho WK, Park S, Shin SW, Park W, Kim H, Choi DH. Checkpoint Kinase 1 (CHK1) Inhibition Enhances the Sensitivity of Triple-Negative Breast Cancer Cells to Proton Irradiation via Rad51 Downregulation. Int J Mol Sci 2020; 21:ijms21082691. [PMID: 32294924 PMCID: PMC7215565 DOI: 10.3390/ijms21082691] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/11/2020] [Accepted: 04/12/2020] [Indexed: 02/07/2023] Open
Abstract
Due to a superior dose conformity to the target, proton beam therapy (PBT) continues to rise in popularity. Recently, considerable efforts have been directed toward discovering treatment options for use in combination with PBT. This study aimed to investigate the targeting of checkpoint kinase 1 (CHK1), a critical player regulating the G2/M checkpoint, as a promising strategy to potentiate PBT in human triple-negative breast cancer (TNBC) cells. Protons induced cell-cycle arrest at the G2/M checkpoint more readily in response to increased CHK1 activation than X-rays. A clonogenic survival assay revealed that CHK1 inhibition using PF-477736 or small interfering RNA (siRNA) enhanced the sensitivity toward protons to a greater extent than toward X-rays. Western blotting demonstrated that PF-477736 treatment in the background of proton irradiation increased the pro-apoptotic signaling, which was further supported by flow cytometry using annexin V. Immunofluorescence revealed that proton-induced DNA double-strand breaks (DSBs) were further enhanced by PF-477736, which was linked to the downregulation of Rad51, essential for the homologous recombination repair of DSBs. Direct inactivation of Rad51 resulted in enhanced proton sensitization. Collectively, these data suggest that targeting CHK1 may be a promising approach for improving PBT efficacy in the treatment of TNBC.
Collapse
Affiliation(s)
- Changhoon Choi
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Korea; (C.C.); (W.K.C.); (S.P.); (S.-W.S.); (W.P.); (H.K.)
| | - Won Kyung Cho
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Korea; (C.C.); (W.K.C.); (S.P.); (S.-W.S.); (W.P.); (H.K.)
| | - Sohee Park
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Korea; (C.C.); (W.K.C.); (S.P.); (S.-W.S.); (W.P.); (H.K.)
| | - Sung-Won Shin
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Korea; (C.C.); (W.K.C.); (S.P.); (S.-W.S.); (W.P.); (H.K.)
- Department of Radiation Oncology, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Won Park
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Korea; (C.C.); (W.K.C.); (S.P.); (S.-W.S.); (W.P.); (H.K.)
- Department of Radiation Oncology, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Haeyoung Kim
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Korea; (C.C.); (W.K.C.); (S.P.); (S.-W.S.); (W.P.); (H.K.)
- Department of Radiation Oncology, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Doo Ho Choi
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Korea; (C.C.); (W.K.C.); (S.P.); (S.-W.S.); (W.P.); (H.K.)
- Department of Radiation Oncology, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Correspondence: ; Tel.: +82-2-3410-2436
| |
Collapse
|
17
|
Xu C, Zhang M, Bian L, Li Y, Yao Y, Li D. N-glycosylated SGK196 suppresses the metastasis of basal-like breast cancer cells. Oncogenesis 2020; 9:4. [PMID: 31913260 PMCID: PMC6949223 DOI: 10.1038/s41389-019-0188-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 01/04/2023] Open
Abstract
SGK196 is a protein O-mannose kinase involved in an indispensable phosphorylation step during laminin-binding glycan synthesis on alpha-dystroglycan (α-DG). However, the function of SGK196 in cancer diseases remains elusive. In the current study, we demonstrated that SGK196 is primarily modified by N-glycosylation in breast cancer (BC) cells. Furthermore, gain and loss-of-function studies showed that N-glycosylated SGK196 suppresses cell migration, invasion, and metastasis in BC, particularly in the basal-like breast cancer (BLBC) type. In addition, we found that SGK196 N-glycosylation performs the regulatory function through the PI3K/AKT/GSK3β signaling pathway. Collectively, our results show that N-glycosylated SGK196 plays suppression roles in BLBC metastases, therefore providing new insights into SGK196 function in BC.
Collapse
Affiliation(s)
- Ci Xu
- Department of Radiation Oncology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Meichao Zhang
- Department of Radiation Oncology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lei Bian
- Department of Radiation Oncology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yanyan Li
- Department of Radiation Oncology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuan Yao
- Department of Radiation Oncology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Dong Li
- Department of Radiation Oncology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
18
|
Garutti M, Pelizzari G, Bartoletti M, Malfatti MC, Gerratana L, Tell G, Puglisi F. Platinum Salts in Patients with Breast Cancer: A Focus on Predictive Factors. Int J Mol Sci 2019; 20:E3390. [PMID: 31295913 PMCID: PMC6678596 DOI: 10.3390/ijms20143390] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/07/2019] [Accepted: 07/08/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC) is the most frequent oncologic cause of death among women and the improvement of its treatments is compelling. Platinum salts (e.g., carboplatin, cisplatin, and oxaliplatin) are old drugs still used to treat BC, especially the triple-negative subgroup. However, only a subset of patients see a concrete benefit from these drugs, raising the question of how to select them properly. Therefore, predictive biomarkers for platinum salts in BC still represent an unmet clinical need. Here, we review clinical and preclinical works in order to summarize the current evidence about predictive or putative platinum salt biomarkers in BC. The association between BRCA1/2 gene mutations and platinum sensitivity has been largely described. However, beyond the mutations of these two genes, several other proteins belonging to the homologous recombination pathways have been linked to platinum response, defining the concept of BRCAness. Several works, here reviewed, have tried to capture BRCAness through different strategies, such as homologous recombination deficiency (HRD) score and genetic signatures. Moreover, p53 and its family members (p63 and p73) might also be used as predictors of platinum response. Finally, we describe the mounting preclinical evidence regarding base excision repair deficiency as a possible new platinum biomarker.
Collapse
Affiliation(s)
- Mattia Garutti
- U.O.C Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Roma, Italy
- Dipartimento di Oncologia Medica, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Giacomo Pelizzari
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
- Dipartimento di Oncologia Medica, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Michele Bartoletti
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
- Dipartimento di Oncologia Medica, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | | | - Lorenzo Gerratana
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
- Dipartimento di Oncologia Medica, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Gianluca Tell
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
| | - Fabio Puglisi
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy.
- Dipartimento di Oncologia Medica, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy.
| |
Collapse
|
19
|
Alexandrou S, George SM, Ormandy CJ, Lim E, Oakes SR, Caldon CE. The Proliferative and Apoptotic Landscape of Basal-like Breast Cancer. Int J Mol Sci 2019; 20:ijms20030667. [PMID: 30720718 PMCID: PMC6387372 DOI: 10.3390/ijms20030667] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 01/21/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023] Open
Abstract
Basal-like breast cancer (BLBC) is an aggressive molecular subtype that represents up to 15% of breast cancers. It occurs in younger patients, and typically shows rapid development of locoregional and distant metastasis, resulting in a relatively high mortality rate. Its defining features are that it is positive for basal cytokeratins and, epidermal growth factor receptor and/or c-Kit. Problematically, it is typically negative for the estrogen receptor and human epidermal growth factor receptor 2 (HER2), which means that it is unsuitable for either hormone therapy or targeted HER2 therapy. As a result, there are few therapeutic options for BLBC, and a major priority is to define molecular subgroups of BLBC that could be targeted therapeutically. In this review, we focus on the highly proliferative and anti-apoptotic phenotype of BLBC with the goal of defining potential therapeutic avenues, which could take advantage of these aspects of tumor development.
Collapse
Affiliation(s)
- Sarah Alexandrou
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 2010 Sydney, Australia.
| | - Sandra Marie George
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 2010 Sydney, Australia.
| | - Christopher John Ormandy
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 2010 Sydney, Australia.
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, 2052 Sydney, Australia.
| | - Elgene Lim
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 2010 Sydney, Australia.
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, 2052 Sydney, Australia.
| | - Samantha Richelle Oakes
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 2010 Sydney, Australia.
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, 2052 Sydney, Australia.
| | - C Elizabeth Caldon
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 2010 Sydney, Australia.
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, 2052 Sydney, Australia.
| |
Collapse
|
20
|
Wang C, Bai F, Zhang LH, Scott A, Li E, Pei XH. Estrogen promotes estrogen receptor negative BRCA1-deficient tumor initiation and progression. Breast Cancer Res 2018; 20:74. [PMID: 29996906 PMCID: PMC6042319 DOI: 10.1186/s13058-018-0996-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/30/2018] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Estrogen promotes breast cancer development and progression mainly through estrogen receptor (ER). However, blockage of estrogen production or action prevents development of and suppresses progression of ER-negative breast cancers. How estrogen promotes ER-negative breast cancer development and progression is poorly understood. We previously discovered that deletion of cell cycle inhibitors p16Ink4a (p16) or p18Ink4c (p18) is required for development of Brca1-deficient basal-like mammary tumors, and that mice lacking p18 develop luminal-type mammary tumors. METHODS A genetic model system with three mouse strains, one that develops ER-positive mammary tumors (p18 single deletion) and the others that develop ER-negative tumors (p16;Brca1 and p18;Brca1 compound deletion), human BRCA1 mutant breast cancer patient-derived xenografts, and human BRCA1-deficient and BRCA1-proficient breast cancer cells were used to determine the role of estrogen in activating epithelial-mesenchymal transition (EMT), stimulating cell proliferation, and promoting ER-negative mammary tumor initiation and metastasis. RESULTS Estrogen stimulated the proliferation and tumor-initiating potential of both ER-positive Brca1-proficient and ER-negative Brca1-deficient tumor cells. Estrogen activated EMT in a subset of Brca1-deficient mammary tumor cells that maintained epithelial features, and enhanced the number of cancer stem cells, promoting tumor progression and metastasis. Estrogen activated EMT independent of ER in Brca1-deficient, but not Brca1-proficient, tumor cells. Estrogen activated the AKT pathway in BRCA1-deficient tumor cells independent of ER, and pharmaceutical inhibition of AKT activity suppressed EMT and cell proliferation preventing BRCA1 deficient tumor progression. CONCLUSIONS This study reveals for the first time that estrogen promotes BRCA1-deficient tumor initiation and progression by stimulation of cell proliferation and activation of EMT, which are dependent on AKT activation and independent of ER.
Collapse
Affiliation(s)
- Chuying Wang
- Department of Medical Oncology, The First Affiliated hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 People’s Republic of China
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136 USA
| | - Feng Bai
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136 USA
| | - Li-han Zhang
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136 USA
| | - Alexandria Scott
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136 USA
| | - Enxiao Li
- Department of Medical Oncology, The First Affiliated hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 People’s Republic of China
| | - Xin-Hai Pei
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136 USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136 USA
| |
Collapse
|
21
|
Park JH, Ahn JH, Kim SB. How shall we treat early triple-negative breast cancer (TNBC): from the current standard to upcoming immuno-molecular strategies. ESMO Open 2018; 3:e000357. [PMID: 29765774 PMCID: PMC5950702 DOI: 10.1136/esmoopen-2018-000357] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2018] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a long-lasting orphan disease in terms of little therapeutic progress during the past several decades and still the standard of care remains chemotherapy. Experimental discovery of molecular signatures including the ‘BRCAness’ highlighted the innate heterogeneity of TNBC, generating the diversity of TNBC phenotypes. As it contributes to enhancing genomic instability, it has widened the therapeutic spectrum of TNBC. In particular, unusual sensitivity to DNA damaging agents was denoted in patients with BRCA deficiency, suggesting therapeutic benefit from platinum and poly(ADP-ribose) polymerase inhibitors. However, regardless of enriched chemosensitivity and immunogenicity, majority of patients with TNBC still suffer from dismal clinical outcomes including early relapse and metastatic spread. Therefore, efforts into more precise and personalised treatment are critical at this point. Accordingly, the advance of multiomics has revealed novel actionable targets including PI3K-Akt-mTOR and epidermal growth factor receptor signalling pathways, which might actively participate in modulating the chemosensitivity and immune system. Also, TNBC has long been considered a potential protagonist of immunotherapy in breast cancer, supported by abundant tumour-infiltrating lymphocytes and heterogeneous tumour microenvironment. Despite that, earlier studies showed somewhat unsatisfactory results of monotherapy with immune-checkpoint inhibitors, consistently durable responses in responders were noteworthy. Based on these results, further combinatorial trials either with other chemotherapy or targeted agents are underway. Incorporating immune-molecular targets into combination as well as refining the standard chemotherapy might be the key to unlock the future of TNBC. In this review, we share the current and upcoming treatment options of TNBC in the framework of scientific and clinical data, especially focusing on early stage of TNBC.
Collapse
Affiliation(s)
- Ji Hyun Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, Korea; Department of Hemato-Oncology, Konkuk Medical Center, University of Konkuk College of Medicine, Gwangjin-gu, Seoul, Korea
| | - Jin-Hee Ahn
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, Korea
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, Korea.
| |
Collapse
|
22
|
Sizemore ST, Mohammad R, Sizemore GM, Nowsheen S, Yu H, Ostrowski MC, Chakravarti A, Xia F. Synthetic Lethality of PARP Inhibition and Ionizing Radiation is p53-dependent. Mol Cancer Res 2018; 16:1092-1102. [PMID: 29592899 DOI: 10.1158/1541-7786.mcr-18-0106] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/09/2018] [Accepted: 03/14/2018] [Indexed: 12/20/2022]
Abstract
PARP inhibitors (PARPi) are potentially effective therapeutic agents capable of inducing synthetic lethality in tumors with deficiencies in homologous recombination (HR)-mediated DNA repair such as those carrying BRCA1 mutations. However, BRCA mutations are rare, the majority of tumors are proficient in HR repair, and thus most tumors are resistant to PARPi. Previously, we observed that ionizing radiation (IR) initiates cytoplasmic translocation of BRCA1 leading to suppression of HR-mediated DNA repair and induction of synthetic PARPi lethality in wild-type BRCA1 and HR-proficient tumor cells. The tumor suppressor p53 was identified as a key factor that regulates DNA damage-induced BRCA1 cytoplasmic sequestration following IR. However, the role of p53 in IR-induced PARPi sensitization remains unclear. This study elucidates the role of p53 in IR-induced PARPi cytotoxicity in HR-proficient cancer cells and suggests p53 status may help define a patient population that might benefit from this treatment strategy. Sensitization to PARPi following IR was determined in vitro and in vivo utilizing human breast and glioma tumor cells carrying wild-type BRCA1 and p53, and in associated cells in which p53 function was modified by knockdown or mutation. In breast and glioma cells with proficient HR repair, IR-induced BRCA1 cytoplasmic sequestration, HR repair inhibition, and subsequent PARPi sensitization in vitro and in vivo was dependent upon functional p53.Implications: Implications: p53 status determines PARP inhibitor sensitization by ionizing radiation in multiple BRCA1 and HR-proficient tumor types and may predict which patients are most likely to benefit from combination therapy. Mol Cancer Res; 16(7); 1092-102. ©2018 AACR.
Collapse
Affiliation(s)
- Steven T Sizemore
- Department of Radiation Oncology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Rahman Mohammad
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Gina M Sizemore
- Department of Radiation Oncology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Clinic, Rochester, Minnesota
| | - Hao Yu
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Michael C Ostrowski
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Arnab Chakravarti
- Department of Radiation Oncology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Fen Xia
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas.
| |
Collapse
|
23
|
miR-151-5p, targeting chromatin remodeler SMARCA5, as a marker for the BRCAness phenotype. Oncotarget 2018; 7:80363-80372. [PMID: 27385001 PMCID: PMC5348325 DOI: 10.18632/oncotarget.10345] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 06/13/2016] [Indexed: 12/12/2022] Open
Abstract
In recent years, the assessment of biomarkers useful for “precision medicine” has been a hot topic in research. The involvement of microRNAs in the pathogenesis of breast cancer has been highly investigated with the aim of being able to molecularly stratify this highly heterogeneous disease. Our aim was to identify microRNAs targeting DNA repair machinery, through Affymetrix GeneChip miRNA Arrays, in a cohort of BRCA-related and sporadic breast cancers. Moreover, we analyzed microRNA expression taking into account our previous results on the expression of PARP1, because of its importance in targeted therapy. miR-361-5p and miR-151-5p were found to be overexpressed in PARP1-upregulating BRCA-germline mutated and sporadic breast tumors. Pathway enrichment analysis was performed to identify potential target genes to be analyzed in the validation step in an independent cohort. Our results confirmed the overexpression of miR-151-5p and, interestingly, its role in the targeting of SMARCA5, a chromatin remodeler. This result was also confirmed in vitro, both through luciferase assay and by analyzing endogenous levels of SMARCA5 in MCF-7 cell lines using miR-151-5p mimic and inhibitor. In conclusion, our data showed the possibility of considering the overexpression of PARP1 and miR-151-5p as biomarkers useful to correctly treat sporadic breast cancers, which eventually could be considered as BRCAness tumors, with PARP-inhibitors.
Collapse
|
24
|
Gregório AC, Lacerda M, Figueiredo P, Simões S, Dias S, Moreira JN. Therapeutic Implications of the Molecular and Immune Landscape of Triple-Negative Breast Cancer. Pathol Oncol Res 2017; 24:701-716. [PMID: 28913723 DOI: 10.1007/s12253-017-0307-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 09/04/2017] [Indexed: 12/15/2022]
Abstract
Treatment and management of breast cancer imposes a heavy burden on public health care, and incidence rates continue to increase. Breast cancer is the most common female neoplasia and primary cause of death among women worldwide. The recognition of breast cancer as a complex and heterogeneous disease, comprising different molecular entities, was a landmark in our understanding of this malignancy. Valuing the impact of the molecular characteristics on tumor behavior enabled a better assessment of a patient's prognosis and increased the predictive power to therapeutic response and clinical outcome. Molecular heterogeneity is also prominent in the triple-negative breast cancer subtype, and is reflected by the distinct prognostic and patient's sensitivity to treatment, being chemotherapy the only systemic treatment currently available. From a therapeutic perspective, gene expression profiling of triple-negative tumors has notably contributed to the exploration of new druggable targets and brought to light the need to align these patients to the various therapies according to their triple-negative subtype. Additionally, the higher amount of tumor infiltrating lymphocytes, and the prevalence of an increased expression of PD-1 receptor and its ligand, PD-L1, in triple-negative tumors, created a new treatment opportunity with immune checkpoint inhibitors. This manuscript addresses the current knowledge on the molecular and immune profiles of breast cancer, and its impact on the development of targeted therapies, with a particular emphasis on the triple-negative subtype.
Collapse
Affiliation(s)
- Ana C Gregório
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine (Pólo I), University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal
- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Manuela Lacerda
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Paulo Figueiredo
- IPOFG-EPE - Portuguese Institute of Oncology Francisco Gentil, Coimbra, Portugal
| | - Sérgio Simões
- FFUC - Faculty of Pharmacy, Pólo das Ciências da Saúde, University of Coimbra, Coimbra, Portugal
| | - Sérgio Dias
- IMM - Institute of Molecular Medicine, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - João Nuno Moreira
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine (Pólo I), University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal.
- FFUC - Faculty of Pharmacy, Pólo das Ciências da Saúde, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
25
|
Abstract
Breast cancer 1 (BRCA1), as a tumor suppressor, exerts an effective influence on protecting DNA integrity to suppress the development of breast cancer (BC). BRCA1 expression is induced in response to DNA-damaging agents such as etoposide. Germline BRCA1 gene mutations are associated with development of hereditary BC. However, besides BRCA-mutated BCs, some sporadic cancers may also exhibit a BRCA-like phenotype, displaying so-called ‘BRCAness’. This common phenotype may respond to similar therapeutic approaches as BRCA-mutated tumors and may thus have important implications for the clinical management of these cancers. In order to determine whether and how etoposide regulates the protein levels of BRCA1 in BC cells, we exposed a panel of five selected cell lines to etoposide, compared the results to untreated control cells, and then stained the cells with the specific, reliable, and reproducible MS110 antibody directed against phosphorylated Ser1423 BRCA1. By evaluating cytoplasmic BRCA1 protein levels, we were able to distinguish three aggressive BC subtypes with BRCAness characteristics. In addition, determination of early and late apoptosis helped to complete the analysis of BRCA1 functions in the DNA damage pathway of aggressive BC. In conclusion, our study suggested that high cytoplasmic BRCA1 protein levels could be considered as a potential predictive marker for response to chemotherapy in both sporadic and hereditary BC. Tumors with either BRCAness phenotype or germline BRCA1 mutation are both aggressive BCs associated with poor prognosis and could both be subjected to targeted therapies against BRCA1-mutated BC in future clinical management strategies.
Collapse
|
26
|
The role of BRCA status on prognosis in patients with triple-negative breast cancer. Oncotarget 2017; 8:87151-87162. [PMID: 29152070 PMCID: PMC5675622 DOI: 10.18632/oncotarget.19895] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 07/18/2017] [Indexed: 02/05/2023] Open
Abstract
Studies have showed that dysfunction in the breast cancer susceptibility gene (BRCA) is associated with triple-negative breast cancer (TNBC); however, its effect on patient survival remains controversial. We investigated the distribution of BRCA1/2 mutations in unselected Chinese patients with TNBC and explored their roles in prognosis. Then a systematic review and meta-analysis were performed to evaluate the prognostic role of BRCA dysfunction, including BRCA1/2 germline/somatic mutations, BRCA1 promoter methylation, and low BRCA1 protein expression in TNBC patients. Pooled hazard ratios with 95% confidence intervals were estimated to determine the association between BRCA dysfunction and survival. Our results showed a high frequency of BRCA1/2 mutations, especially germline BRCA1 variants, were associated with bilateral breast cancer. Although no correlations were found between BRCA1/2 mutations and recurrence-free survival (RFS) or overall survival (OS). In the meta-analysis, patients with BRCA1 promoter methylation showed poor OS. However, there was a favorable impact on disease free survival (DFS) for TNBC patients with BRCA1 promoter methylation when received adjuvant-chemotherapy. In conclusion, BRCA1/2 mutations were associated with bilateral breast cancer and BRCA1 promoter methylation may have a prognostic effect on TNBC.
Collapse
|
27
|
Caruso D, Papa A, Tomao S, Vici P, Panici PB, Tomao F. Niraparib in ovarian cancer: results to date and clinical potential. Ther Adv Med Oncol 2017; 9:579-588. [PMID: 29081841 PMCID: PMC5564880 DOI: 10.1177/1758834017718775] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 06/09/2017] [Indexed: 12/26/2022] Open
Abstract
Ovarian cancer is the first cause of death from gynaecological malignancy. Germline mutation in BRCA1 and 2, two genes involved in the mechanisms of reparation of DNA damage, are showed to be related with the incidence of breast and ovarian cancer, both sporadic and familiar. PARP is a family of enzymes involved in the base excision repair (BER) system. The introduction of inhibitors of PARP in patients with BRCA-mutated ovarian cancer is correlated with the concept of synthetic lethality. Among the PARP inhibitors introduced in clinical practice, niraparib showed interesting results in a phase III trial in the setting of maintenance treatment in ovarian cancer, after platinum-based chemotherapy. Interestingly, was niraparib showed to be efficacious not only in BRCA-mutated patients, but also in patients with other alterations of the homologous recombination (HR) system and in patients with unknown alterations. These results position niraparib as the first PARP-inhibitor with clinically and statistically significant results also in patients with no alterations in BRCA 1/2 and other genes involved in the DNA repair system. Even if the results are potentially practice-changing, the action of niraparib must be further studied and deepened.
Collapse
Affiliation(s)
- Davide Caruso
- Department of Medico-Surgical Sciences and Biotechnologies, University of Rome 'Sapienza', Latina, Italy
| | - Anselmo Papa
- Department of Medico-Surgical Sciences and Biotechnologies, University of Rome 'Sapienza', Corso della Repubblica 79, 04100, Latina, Italy
| | - Silverio Tomao
- Department of Medico-Surgical Sciences and Biotechnologies, University of Rome 'Sapienza', Latina, Italy
| | - Patrizia Vici
- Division of Medical Oncology 2, 'Regina Elena' National Cancer Institute, Rome, Italy
| | | | - Federica Tomao
- Department of Gynaecology and Obstetrics, University of Rome 'Sapienza', Rome, Italy; Department of Gynecology, University of Heraklion, Heraklion, Greece
| |
Collapse
|
28
|
Jitariu AA, Cîmpean AM, Ribatti D, Raica M. Triple negative breast cancer: the kiss of death. Oncotarget 2017; 8:46652-46662. [PMID: 28445140 PMCID: PMC5542300 DOI: 10.18632/oncotarget.16938] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/19/2017] [Indexed: 12/22/2022] Open
Abstract
One of the most controversial women malignancies, triple negative breast cancers (TNBCs) are critically overviewed here, being focused on data useful in clinical practice or to improve the therapy and patients survival. TNBCs "choose" young women and its "kiss" is, unfortunately deadly in most cases. Currently, few sparse data are available in literature concerning the origins of TNBC. Vasculogenic mimicry detected in TNBCs, seems to be determined by a population of CD133+ cells and may be stimulated by different pharmacological agents such sunitinib. Despite the fact that TNBCs do not usually metastasize through the lymphatic pathways, TNBCs may be characterized by lymphatic invasion and by an increased lymphatic microvascular density. If TNBCs treatment depends on the molecular profile of the tumor, the same statement may be postulated for TNBCs metastasis. Whether metastases have a similar phenotype as the primary tumor remains an enigma. Therefore, the question: 'Could TNBC be subject to a standardized, unanimously accepted therapeutic strategy or is it strictly subclass-dependent?' remains to be further investigated.
Collapse
Affiliation(s)
- Adriana-Andreea Jitariu
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| | - Anca Maria Cîmpean
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
- National Cancer Institute “ Giovanni Paolo II”, Bari, Italy
| | - Marius Raica
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| |
Collapse
|
29
|
Burke AJ, Garrido P, Johnson C, Sullivan FJ, Glynn SA. Inflammation and Nitrosative Stress Effects in Ovarian and Prostate Pathology and Carcinogenesis. Antioxid Redox Signal 2017; 26:1078-1090. [PMID: 28326819 DOI: 10.1089/ars.2017.7004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
SIGNIFICANCE Prostate and ovarian cancers are major contributors to cancer-related deaths worldwide. Recently, inflammation and nitrosative stress have been implicated in carcinogenesis, with the overexpression of NOS2 and concomitant release of nitric oxide (NO) associated with cancer initiation and progression. Recent Advances: An increasing body of evidence indicates an association between NOS2 expression and aggressive ovarian cancer. Research also indicates a role for NO in prostate disease pathology and prostate cancer. A therapeutic role for NOS2 inhibition and/or NO drugs exists for the treatment of both ovarian and prostate tumors. CRITICAL ISSUES Herein, we review the key molecular effects associated with NOS2 in ovarian and prostate cancer. NOS2 increases angiogenesis and tumor proliferation and correlates with aggressive type II ovarian tumors. NOS2 expressing tumors are sensitive to cisplatin chemotherapy, and NO may be used to sensitize cisplatin-resistant tumors to chemotherapy. NOS2 is highly expressed in prostate tumors compared to non-neoplastic prostate pathologies. NO may play a role in the development of androgen-independent prostate cancer via s-nitrosylation of the androgen receptor. Moreover, NOS2 inhibitors and NO donor drugs show therapeutic potential in ovarian and prostate cancer as single agents or dual drugs, by either inhibiting the effects of NOS2 or increasing NO levels to induce cytotoxic effects. FUTURE DIRECTIONS NOS2 and NO present new targets for the treatment of ovarian and prostate tumors. Furthermore, understanding NO-related tumor biology in these cancers presents a new means for improved patient stratification to the appropriate treatment regimen. Antioxid. Redox Signal. 26, 1078-1090.
Collapse
Affiliation(s)
- Amy J Burke
- 1 Prostate Cancer Institute, School of Medicine, National University of Ireland Galway , Galway, Ireland
| | - Pablo Garrido
- 1 Prostate Cancer Institute, School of Medicine, National University of Ireland Galway , Galway, Ireland .,2 Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway , Galway, Ireland
| | - Carol Johnson
- 1 Prostate Cancer Institute, School of Medicine, National University of Ireland Galway , Galway, Ireland .,2 Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway , Galway, Ireland
| | - Francis J Sullivan
- 1 Prostate Cancer Institute, School of Medicine, National University of Ireland Galway , Galway, Ireland
| | - Sharon A Glynn
- 1 Prostate Cancer Institute, School of Medicine, National University of Ireland Galway , Galway, Ireland .,2 Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway , Galway, Ireland .,3 Apoptosis Research Centre, Biomedical Sciences, National University of Ireland Galway , Galway, Ireland
| |
Collapse
|
30
|
Jin J, Zhang W, Ji W, Yang F, Guan X. Predictive biomarkers for triple negative breast cancer treated with platinum-based chemotherapy. Cancer Biol Ther 2017; 18:369-378. [PMID: 28494179 DOI: 10.1080/15384047.2017.1323582] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Treatment of triple negative breast cancer (TNBC) has been a big challenge since it is defined. To date, platinum-based chemotherapy has played a significant role in the treatment of TNBC patients. However, some patients do not respond to platinum salts or gradually develop chemoresistance, resulting in little effect, or even some adverse effects. Here, we review numerous preclinical and clinical investigations to summarize possible mechanisms and potential predictive biomarkers of platinum in TNBC. The homologous recombination deficiency (HRD) resulting from the loss of BRCA function is the main rationale of platinum efficacy in TNBC. BRCA mutation and methylation have been demonstrated to be important potential biomarkers. Based on genome-wide effects, BRCA-like classifier can identify the functional loss of BRCA and work as the predictor. HRD score that is able to identify the "BRCAness" and predict the sensitivity of platinum is increasingly considered. Taken together, all findings suggest that HR deficiency profile encompassed by BRCA mutation and high HRD score could predict response to platinum, even to other DNA-damage inducing agents. p53 family members and molecular subtypes of TNBC are also important alternative considerations for predicting platinum response based on the preclinical trials. Currently, tumor infiltrating lymphocyte level and thrombocytopenia are emerging as predictive biomarkers.
Collapse
Affiliation(s)
- Juan Jin
- a Department of Medical Oncology , Jinling Hospital, Medical School of Nanjing University , Nanjing , China
| | - Wenwen Zhang
- a Department of Medical Oncology , Jinling Hospital, Medical School of Nanjing University , Nanjing , China
| | - Wenfei Ji
- b Department of Medical Oncology , Jinling Hospital, Medical School of Nanjing Medical University , Nanjing , China
| | - Fang Yang
- a Department of Medical Oncology , Jinling Hospital, Medical School of Nanjing University , Nanjing , China
| | - Xiaoxiang Guan
- a Department of Medical Oncology , Jinling Hospital, Medical School of Nanjing University , Nanjing , China.,b Department of Medical Oncology , Jinling Hospital, Medical School of Nanjing Medical University , Nanjing , China
| |
Collapse
|
31
|
Domagala P, Hybiak J, Cybulski C, Lubinski J. BRCA1/2-negative hereditary triple-negative breast cancers exhibit BRCAness. Int J Cancer 2017; 140:1545-1550. [DOI: 10.1002/ijc.30570] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 10/30/2016] [Accepted: 11/30/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Pawel Domagala
- Department of Pathology; Pomeranian Medical University; Szczecin Poland
| | - Jolanta Hybiak
- Department of Pathology; Pomeranian Medical University; Szczecin Poland
| | - Cezary Cybulski
- Department of Genetics and Pathology; International Hereditary Cancer Center, Pomeranian Medical University; Szczecin Poland
| | - Jan Lubinski
- Department of Genetics and Pathology; International Hereditary Cancer Center, Pomeranian Medical University; Szczecin Poland
| |
Collapse
|
32
|
Silwal-Pandit L, Langerød A, Børresen-Dale AL. TP53 Mutations in Breast and Ovarian Cancer. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a026252. [PMID: 27815305 DOI: 10.1101/cshperspect.a026252] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Breast and ovarian cancers are the second and fifth leading causes of cancer deaths among women. Both breast and ovarian cancers are highly heterogeneous and are presented with diverse morphology, natural history, and response to therapy. In recent years, international efforts have led to extensive molecular characterization of both breast and ovarian tumors and identified biologically and clinically relevant subtypes of the diseases based on these molecular features. The role of TP53 in tumor initiation and progression is context dependent, and abrogation of the TP53 pathway seems to be essential for the development of basal-like breast cancers and high-grade serous ovarian cancers. These subtypes of breast and ovarian cancer show several genomic similarities including high frequency of TP53 mutation, which seems to be an early, initiating, and driving alteration in these cancer subtypes.
Collapse
Affiliation(s)
- Laxmi Silwal-Pandit
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital The Norwegian Radium Hospital, Montebello, 0310 Oslo, Norway
| | - Anita Langerød
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital The Norwegian Radium Hospital, Montebello, 0310 Oslo, Norway
| | - Anne-Lise Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital The Norwegian Radium Hospital, Montebello, 0310 Oslo, Norway
| |
Collapse
|
33
|
DNA damage repair in breast cancer and its therapeutic implications. Pathology 2016; 49:156-165. [PMID: 28034453 DOI: 10.1016/j.pathol.2016.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/06/2016] [Accepted: 11/02/2016] [Indexed: 11/23/2022]
Abstract
The DNA damage response (DDR) involves the activation of numerous cellular activities that repair DNA lesions and maintain genomic integrity, and is critical in preventing tumorigenesis. Inherited or acquired mutations in specific genes involved in the DNA damage response, for example the breast cancer susceptibility genes 1/2 (BRCA1/2), phosphatase and tensin homolog (PTEN) and P53 are associated with various subtypes of breast cancer. Such changes can render breast cancer cells particularly sensitive to specific DNA damage response inhibitors, for example BRCA1/2 germline mutated cells are sensitive to poly (ADP-ribose) polymerase (PARP) inhibitors. The aims of this review are to discuss specific DNA damage response defects in breast cancer and to present the current stage of development of various DDR inhibitors (namely PARP, ATM/ATR, DNA-PK, PARG, RECQL5, FEN1 and APE1) for breast cancer mono- and combination therapy.
Collapse
|
34
|
Scott A, Bai F, Chan HL, Liu S, Ma J, Slingerland JM, Robbins DJ, Capobianco AJ, Pei XH. p16INK4a suppresses BRCA1-deficient mammary tumorigenesis. Oncotarget 2016; 7:84496-84507. [PMID: 27811360 PMCID: PMC5356676 DOI: 10.18632/oncotarget.13015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/25/2016] [Indexed: 11/25/2022] Open
Abstract
Senescence prevents the proliferation of genomically damaged, but otherwise replication competent cells at risk of neoplastic transformation. p16INK4A (p16), an inhibitor of CDK4 and CDK6, plays a critical role in controlling cellular senescence in multiple organs. Functional inactivation of p16 by gene mutation and promoter methylation is frequently detected in human breast cancers. However, deleting p16 in mice or targeting DNA methylation within the murine p16 promoter does not result in mammary tumorigenesis. How loss of p16 contributes to mammary tumorigenesis in vivo is not fully understood.In this article, we reported that disruption of Brca1 in the mammary epithelium resulted in premature senescence that was rescued by p16 loss. We found that p16 loss transformed Brca1-deficient mammary epithelial cells and induced mammary tumors, though p16 loss alone was not sufficient to induce mammary tumorigenesis. We demonstrated that loss of both p16 and Brca1 led to metastatic, basal-like, mammary tumors with the induction of EMT and an enrichment of tumor initiating cells. We discovered that promoter methylation silenced p16 expression in most of the tumors developed in mice heterozygous for p16 and lacking Brca1. These data not only identified the function of p16 in suppressing BRCA1-deficient mammary tumorigenesis, but also revealed a collaborative effect of genetic mutation of p16 and epigenetic silencing of its transcription in promoting tumorigenesis. To the best of our knowledge, this is the first genetic evidence directly showing that p16 which is frequently deleted and inactivated in human breast cancers, collaborates with Brca1 controlling mammary tumorigenesis.
Collapse
MESH Headings
- Animals
- BRCA1 Protein/genetics
- BRCA1 Protein/metabolism
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cells, Cultured
- Cyclin-Dependent Kinase Inhibitor p16/genetics
- Cyclin-Dependent Kinase Inhibitor p16/metabolism
- DNA Methylation
- Epithelial Cells/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/pathology
- Mice, Knockout
- Mice, Transgenic
- Promoter Regions, Genetic/genetics
Collapse
Affiliation(s)
- Alexandria Scott
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Feng Bai
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Ho Lam Chan
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Shiqin Liu
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Jinshan Ma
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Joyce M Slingerland
- Braman Family Breast Cancer Institute, Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - David J. Robbins
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Anthony J. Capobianco
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Xin-Hai Pei
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
35
|
Badoer C, Garrec C, Goossens D, Ellison G, Mills J, Dzial M, Housni HE, Berwouts S, Concolino P, Guevellou VGL, Delnatte C, Favero JD, Capoluongo E, Bézieau S. Performance of multiplicom's BRCA MASTR Dx kit on the detection of BRCA1 and BRCA2 mutations in fresh frozen ovarian and breast tumor samples. Oncotarget 2016; 7:81357-81366. [PMID: 27793035 PMCID: PMC5348397 DOI: 10.18632/oncotarget.12877] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 09/08/2016] [Indexed: 12/25/2022] Open
Abstract
Next-generation sequencing (NGS) has enabled new approaches for detection of mutations in the BRCA1 and BRCA2 genes responsible for hereditary breast and ovarian cancer (HBOC). The search for germline mutations in the BRCA1 and BRCA2 genes is of importance with respect to oncogenetic and surgical (bilateral mastectomy, ovariectomy) counselling. Testing tumor material for BRCA mutations is of increasing importance for therapeutic decision making as the poly ADP ribose polymerase (PARP) inhibitor, olaparib, is now available to treat patients with specific forms of ovarian cancer and BRCA mutations. Molecular genetics laboratories should develop reliable and sensitive techniques for the complete analysis of the BRCA1 and BRCA2 genes. This is a challenge due to the size of the coding sequence of the BRCA1/2 genes, the absence of hot spot mutations, and particularly by the lower DNA quality obtained from Formalin-Fixed Paraffin-Embedded (FFPE) tissue. As a result, a number of analyses are uninterpretable and do not always provide a result to the clinician, limiting the optimal therapeutic management of patients. The availability of Fresh Frozen Tissue (FFT) for some laboratories and the excellent quality of the DNA extracted from it offers an alternative. For this reason, we evaluated Multiplicom's BRCA MASTR Dx assay on a set of 97 FFT derived DNA samples, in combination with the MID for Illumina MiSeq for BRCA1 and BRCA2 mutation detection. We obtained interpretable NGS results for all tested samples and showed > 99,7% sensitivity, specificity and accuracy.
Collapse
Affiliation(s)
- Cindy Badoer
- Laboratoire de Génétique Moléculaire, Clinique Universitaire de Bruxelles-Hôpital Erasme-Université Libre de Bruxelles (CUB-Erasme-ULB), Brussels, Belgium
| | - Céline Garrec
- Institut de Biologie, Laboratoire de Génétique Moléculaire, Service de Génétique Médicale, CHU Nantes, Nantes, France
| | | | - Gillian Ellison
- AstraZeneca, Personalised Healthcare and Biomarkers, Alderley Park, Macclesfield, UK
| | - John Mills
- AstraZeneca, Personalised Healthcare and Biomarkers, Alderley Park, Macclesfield, UK
| | - Mélina Dzial
- Laboratoire de Génétique Moléculaire, Clinique Universitaire de Bruxelles-Hôpital Erasme-Université Libre de Bruxelles (CUB-Erasme-ULB), Brussels, Belgium
| | - Hakim El Housni
- Laboratoire de Génétique Moléculaire, Clinique Universitaire de Bruxelles-Hôpital Erasme-Université Libre de Bruxelles (CUB-Erasme-ULB), Brussels, Belgium
| | | | - Paola Concolino
- Laboratory of Clinical Molecular and Personalized Diagnostics, Foundation Policlinico Gemelli and Catholic University of Rome, Italy
| | - Virginie Guibert-Le Guevellou
- Institut de Biologie, Laboratoire de Génétique Moléculaire, Service de Génétique Médicale, CHU Nantes, Nantes, France
| | - Capucine Delnatte
- Institut de Biologie, Laboratoire de Génétique Moléculaire, Service de Génétique Médicale, CHU Nantes, Nantes, France
| | | | - Ettore Capoluongo
- Laboratory of Clinical Molecular and Personalized Diagnostics, Foundation Policlinico Gemelli and Catholic University of Rome, Italy
- Molipharma and Giovanni Paolo II Foundation, Campobasso, Italy
| | - Stéphane Bézieau
- Institut de Biologie, Laboratoire de Génétique Moléculaire, Service de Génétique Médicale, CHU Nantes, Nantes, France
| |
Collapse
|
36
|
Cava C, Colaprico A, Bertoli G, Bontempi G, Mauri G, Castiglioni I. How interacting pathways are regulated by miRNAs in breast cancer subtypes. BMC Bioinformatics 2016; 17:348. [PMID: 28185585 PMCID: PMC5123339 DOI: 10.1186/s12859-016-1196-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND An important challenge in cancer biology is to understand the complex aspects of the disease. It is increasingly evident that genes are not isolated from each other and the comprehension of how different genes are related to each other could explain biological mechanisms causing diseases. Biological pathways are important tools to reveal gene interaction and reduce the large number of genes to be studied by partitioning it into smaller paths. Furthermore, recent scientific evidence has proven that a combination of pathways, instead than a single element of the pathway or a single pathway, could be responsible for pathological changes in a cell. RESULTS In this paper we develop a new method that can reveal miRNAs able to regulate, in a coordinated way, networks of gene pathways. We applied the method to subtypes of breast cancer. The basic idea is the identification of pathways significantly enriched with differentially expressed genes among the different breast cancer subtypes and normal tissue. Looking at the pairs of pathways that were found to be functionally related, we created a network of dependent pathways and we focused on identifying miRNAs that could act as miRNA drivers in a coordinated regulation process. CONCLUSIONS Our approach enables miRNAs identification that could have an important role in the development of breast cancer.
Collapse
Affiliation(s)
- Claudia Cava
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Milan, Italy
| | - Antonio Colaprico
- Interuniversity Institute of Bioinformatics in Brussels (IB), Brussels, Belgium
- Machine Learning Group, ULB, Brussels, Belgium
| | - Gloria Bertoli
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Milan, Italy
| | - Gianluca Bontempi
- Interuniversity Institute of Bioinformatics in Brussels (IB), Brussels, Belgium
- Machine Learning Group, ULB, Brussels, Belgium
| | - Giancarlo Mauri
- Department of Informatics, Systems and Communications, University of Milan–Bicocca, Milan, Italy
| | - Isabella Castiglioni
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Milan, Italy
| |
Collapse
|
37
|
Wang Y, Mark KMK, Ung MH, Kettenbach A, Miller T, Xu W, Cheng W, Xia T, Cheng C. Application of RNAi-induced gene expression profiles for prognostic prediction in breast cancer. Genome Med 2016; 8:114. [PMID: 27788678 PMCID: PMC5084341 DOI: 10.1186/s13073-016-0363-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 10/10/2016] [Indexed: 12/17/2022] Open
Abstract
Homologous recombination (HR) is the primary pathway for repairing double-strand DNA breaks implicating in the development of cancer. RNAi-based knockdowns of BRCA1 and RAD51 in this pathway have been performed to investigate the resulting transcriptomic profiles. Here we propose a computational framework to utilize these profiles to calculate a score, named RNA-Interference derived Proliferation Score (RIPS), which reflects cell proliferation ability in individual breast tumors. RIPS is predictive of breast cancer classes, prognosis, genome instability, and neoadjuvant chemosensitivity. This framework directly translates the readout of knockdown experiments into potential clinical applications and generates a robust biomarker in breast cancer.
Collapse
Affiliation(s)
- Yue Wang
- School of Electronic Information and Communications, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China.,Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Kenneth M K Mark
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Matthew H Ung
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Arminja Kettenbach
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, 03766, USA.,Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Todd Miller
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA.,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, 03766, USA
| | - Wei Xu
- School of Electronic Information and Communications, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Wenqing Cheng
- School of Electronic Information and Communications, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Tian Xia
- School of Electronic Information and Communications, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China.
| | - Chao Cheng
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA. .,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, 03766, USA. .,Department of Biomedical Data Sciences, Geisel School of Medicine at Dartmouth, Lebanon, NH, 03766, USA.
| |
Collapse
|
38
|
Iyevleva AG, Imyanitov EN. Cytotoxic and targeted therapy for hereditary cancers. Hered Cancer Clin Pract 2016; 14:17. [PMID: 27555886 PMCID: PMC4994296 DOI: 10.1186/s13053-016-0057-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 06/27/2016] [Indexed: 12/21/2022] Open
Abstract
There is a number of drugs demonstrating specific activity towards hereditary cancers. For example, tumors in BRCA1/2 mutation carriers usually arise via somatic inactivation of the remaining BRCA allele, which makes them particularly sensitive to platinum-based drugs, PARP inhibitors (PARPi), mitomycin C, liposomal doxorubicin, etc. There are several molecular assays for BRCA-ness, which permit to reveal BRCA-like phenocopies among sporadic tumors and thus extend clinical indications for the use of BRCA-specific therapies. Retrospective data on high-dose chemotherapy deserve consideration given some unexpected instances of cure from metastatic disease among BRCA1/2-mutated patients. Hereditary non-polyposis colorectal cancer (HNPCC) is characterized by high-level microsatellite instability (MSI-H), increased antigenicity and elevated expression of immunosuppressive molecules. Recent clinical trial demonstrated tumor responses in HNPCC patients treated by the immune checkpoint inhibitor pembrolizumab. There are successful clinical trials on the use of novel targeted agents for the treatment or rare cancer syndromes, e.g. RET inhibitors for hereditary medullary thyroid cancer, mTOR inhibitors for tumors arising in patients with tuberous sclerosis (TSC), and SMO inhibitors for basal-cell nevus syndrome. Germ-line mutation tests will be increasingly used in the future for the choice of the optimal therapy, therefore turnaround time for these laboratory procedures needs to be significantly reduced to ensure proper treatment planning.
Collapse
Affiliation(s)
- Aglaya G Iyevleva
- N.N. Petrov Institute of Oncology, Pesochny-2, St. Petersburg, 197758 Russia ; St. Petersburg Pediatric Medical University, St. Petersburg, 194100 Russia
| | - Evgeny N Imyanitov
- N.N. Petrov Institute of Oncology, Pesochny-2, St. Petersburg, 197758 Russia ; St. Petersburg Pediatric Medical University, St. Petersburg, 194100 Russia ; I.I. Mechnikov North-Western Medical University, St. Petersburg, 191015 Russia ; St. Petersburg State University, St. Petersburg, 199034 Russia
| |
Collapse
|
39
|
Anderson RC, Makvandi M, Xu K, Lieberman BP, Zeng C, Pryma DA, Mach RH. Iodinated benzimidazole PARP radiotracer for evaluating PARP1/2 expression in vitro and in vivo. Nucl Med Biol 2016; 43:752-758. [PMID: 27689533 DOI: 10.1016/j.nucmedbio.2016.08.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 07/28/2016] [Accepted: 08/08/2016] [Indexed: 01/25/2023]
Abstract
BACKGROUND PARP inhibitors (PARPi) have the potential to impact cancer therapy in a selective patient population; however, despite current patient selection methods clinical trials have shown mixed response rates. It is therefore clinically useful to determine which patients will respond prior to receiving PARPi therapy. One essential biomarker is to measure the level of PARP enzyme expression in tumors. Small molecule radiotracers have been developed to accurately quantify PARP-1 expression in vitro and in vivo. [125I]KX-02-019 is the first report of a radioiodinated analogue of the benzimidazole class of PARPi. Herein, we studied the pharmacological properties of [125I]KX-02-019 as well as the in vivo biodistribution. METHODS [125I]KX-02-019 was evaluated in both cancer and non-cancer cell lines. We evaluated the pharmacologic properties of [125I]KX-02-019 in live cells by measuring enzyme association and dissociation kinetics, saturation, and specificity. In addition, competitive inhibition experiments were carried out with commercially available PARPi. Protein expression was analyzed by Western blot to compare PARP-1 and PARP-2 expression across cell lines studied. The biodistribution was studied in a mouse EMT6 tumor model at time points of 0.5, 1, 2, 4 and 6h. RESULTS [125I]KX-02-019 showed subtle differences in pharmacological properties in the absence of PARP-2. In addition, [125I]KX-02-019 was competitively displaced by clinical PARPi. In vivo biodistribution studies showed an increasing tumor to muscle ratio over 6h as well as fast clearance from healthy tissues. CONCLUSION [125I]KX-02-019 has binding sites in both PARP1 KO cells as well as PARP2 KO cells showing higher affinity for PARP-2. This observation is supported by a decrease in binding affinity in PARP2 KO cells compared to PARP1 KO cells. The pharmacologic and biological properties of [125I]KX-02-019 studied in vitro and in vivo showed that this analogue may be useful in determining pharmacokinetic and pharmacodynamic properties of clinical PARPi.
Collapse
Affiliation(s)
- Redmond-Craig Anderson
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology and Division of Nuclear Medicine and Clinical Molecular Imaging, Philadelphia, PA, 19104, USA
| | - Mehran Makvandi
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology and Division of Nuclear Medicine and Clinical Molecular Imaging, Philadelphia, PA, 19104, USA
| | - Kuiying Xu
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology and Division of Nuclear Medicine and Clinical Molecular Imaging, Philadelphia, PA, 19104, USA
| | - Brian P Lieberman
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology and Division of Nuclear Medicine and Clinical Molecular Imaging, Philadelphia, PA, 19104, USA
| | - Chenbo Zeng
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology and Division of Nuclear Medicine and Clinical Molecular Imaging, Philadelphia, PA, 19104, USA
| | - Daniel A Pryma
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology and Division of Nuclear Medicine and Clinical Molecular Imaging, Philadelphia, PA, 19104, USA
| | - Robert H Mach
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology and Division of Nuclear Medicine and Clinical Molecular Imaging, Philadelphia, PA, 19104, USA.
| |
Collapse
|
40
|
Abdel-Fatah TMA, Arora A, Moseley PM, Perry C, Rakha EA, Green AR, Chan SYT, Ellis IO, Madhusudan S. DNA repair prognostic index modelling reveals an essential role for base excision repair in influencing clinical outcomes in ER negative and triple negative breast cancers. Oncotarget 2016; 6:21964-78. [PMID: 26267318 PMCID: PMC4673139 DOI: 10.18632/oncotarget.4157] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 05/20/2015] [Indexed: 01/23/2023] Open
Abstract
Stratification of oestrogen receptor (ER) negative and triple negative breast cancers (TNBCs) is urgently needed. In the current study, a cohort of 880 ER- (including 635 TNBCs) was immuno-profiled for a panel of DNA repair proteins including: Pol β, FEN1, APE1, XRCC1, SMUG1, PARP1, BRCA1, ATR, ATM, DNA-PKcs, Chk1, Chk2, p53, and TOPO2. Multivariate Cox proportional hazards models (with backward stepwise exclusion of these factors, using a criterion of p < 0.05 for retention of factors in the model) were used to identify factors that were independently associated with clinical outcomes. XRCC1 (p = 0.002), pol β (p = 0.032) FEN1 (p = 0.001) and BRCA1 (p = 0.040) levels were independently associated with poor BCSS. Subsequently, DNA repair index prognostic (DRPI) scores for breast cancer specific survival (BCSS) were calculated and two prognostic groups (DRPI-PGs) were identified. Patients in prognostic group 2 (DRPI-PG2) have higher risk of death (p < 0.001). Furthermore, in DRPI-PG2 patients, exposure to anthracycline reduced the risk of death [(HR (95% CI) = 0.79 (0.64–0.98), p = 0.032) by 21–26%. In addition, DRPI-PG2 patients have adverse clinicopathological features including higher grade, lympho-vascular invasion, Her-2 positive phenotype, compared to those in DRPI-PG1 (p < 0.01). Receiver operating characteristic (ROC) curves indicated that the DRPI outperformed the currently used prognostic factors and adding DRPI to lymph node stage significantly improved their performance as a predictor for BCSS [p < 0.00001, area under curve (AUC) = 0.70]. BER strongly influences pathogenesis of ER- and TNBCs. The DRPI accurately predicts BCSS and can also serve as a valuable prognostic and predictive tool for TNBCs.
Collapse
Affiliation(s)
| | - Arvind Arora
- Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG51 PB, UK
| | - Paul M Moseley
- Department of Oncology, Nottingham University Hospitals, Nottingham NG5 1PB, UK
| | - Christina Perry
- Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG51 PB, UK
| | - Emad A Rakha
- Department of Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
| | - Andrew R Green
- Department of Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
| | - Stephen Y T Chan
- Department of Oncology, Nottingham University Hospitals, Nottingham NG5 1PB, UK
| | - Ian O Ellis
- Department of Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
| | - Srinivasan Madhusudan
- Department of Oncology, Nottingham University Hospitals, Nottingham NG5 1PB, UK.,Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG51 PB, UK
| |
Collapse
|
41
|
Abstract
Autoantibodies reactive against host DNA are detectable in the circulation of most people with systemic lupus erythematosus (SLE). The long-held view that antibodies cannot penetrate live cells has been disproved. A subset of lupus autoantibodies penetrate cells, translocate to nuclei, and inhibit DNA repair or directly damages DNA. The result of these effects depends on the microenvironment and genetic traits of the cell. Some DNA-damaging antibodies alone have little impact on normal cells, but in the presence of other conditions, such as pre-existing DNA-repair defects, can become highly toxic. These findings raise new questions about autoimmunity and DNA damage, and reveal opportunities for new targeted therapies against malignancies particularly vulnerable to DNA damage. In this Perspectives article, we review the known associations between SLE, DNA damage and cancer, and propose a theory for the effects of DNA-damaging autoantibodies on SLE pathophysiology and cancer risk.
Collapse
|
42
|
Dizdar O, Arslan C, Altundag K. Advances in PARP inhibitors for the treatment of breast cancer. Expert Opin Pharmacother 2015; 16:2751-8. [PMID: 26485111 DOI: 10.1517/14656566.2015.1100168] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Poly(ADP-Ribose) polymerases (PARPs) are one of the important components of base excision repair pathway for single strand DNA breaks. Currently accepted hypothesis for the mechanism of action for PARP inhibitors in tumors with homologous recombination deficiency is synthetic lethality, as the simultaneous blockage of both pathways prevents the tumor cells from repairing DNA damage. Other proposed mechanisms include PARP trapping, defective BRCA1 and POLQ recruitment to sites of DNA repair. Breast cancer subgroups with germline BRCA mutations or non-mutational functional defects in BRCA proteins exemplify potential targets for PARP inhibitors. AREAS COVERED Promising results have been achieved with PARP inhibitors in BRCA associated cancers, particularly in ovarian and breast cancer. Olaparib is the only PARP inhibitor approved by FDA in the treatment of patients with germline BRCA mutated advanced ovarian cancer pretreated with ≥3 prior lines of chemotherapy. In this article, we reviewed the current status of PARP inhibitors, completed and ongoing trials, safety and resistance issues in patients with breast cancer. EXPERT OPINION PARP inhibitors show promise in cancers with BRCA mutation and in the treatment of sporadic cancers with defective homologous recombination. Predictors of response, strategies to overcome resistance, combination with other chemotherapies and targeted agents, optimum dose and schedule of administration should be investigated in future trials.
Collapse
Affiliation(s)
- Omer Dizdar
- a Department of Preventive Oncology , Hacettepe University Institute of Cancer , Ankara 06100 , Turkey
| | - Cagatay Arslan
- b Department of Medical Oncology , Izmir University Faculty of Medicine , Izmir 35540 , Turkey
| | - Kadri Altundag
- c Department of Medical Oncology , Hacettepe University Institute of Cancer , Ankara 06100 , Turkey
| |
Collapse
|
43
|
Liang BY, Xiong M, Ji GB, Zhang EL, Zhang ZY, Dong KS, Chen XP, Huang ZY. Synergistic suppressive effect of PARP-1 inhibitor PJ34 and HDAC inhibitor SAHA on proliferation of liver cancer cells. JOURNAL OF HUAZHONG UNIVERSITY OF SCIENCE AND TECHNOLOGY. MEDICAL SCIENCES = HUA ZHONG KE JI DA XUE XUE BAO. YI XUE YING DE WEN BAN = HUAZHONG KEJI DAXUE XUEBAO. YIXUE YINGDEWEN BAN 2015; 35:535-540. [PMID: 26223923 DOI: 10.1007/s11596-015-1466-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 05/26/2015] [Indexed: 10/23/2022]
Abstract
Poly (ADP-ribose) polymerase-1 (PARP-1) inhibitors and histone deacetylase (HDAC) inhibitors have recently emerged as promising anticancer drugs. The aim of this study was to investigate the effect of combination treatment with the PARP inhibitor PJ34 and HDAC inhibitor SAHA on the proliferation of liver cancer cells. Cell proliferation and apoptosis were assessed in three human liver cancer cell lines (HepG2, Hep3B and HCC-LM3) treated with PJ34 (8 μmol/L) and SAHA (1 μmol/L), alone or combined, by Cell Counting Kit-8 assay and flow cytometry, respectively. The nude mice bearing subcutaneous HepG2 tumors were administered different groups of drugs (10 mg/kg PJ34, 25 mg/kg SAHA, 10 mg/kg PJ34+25 mg/kg SAHA), and the inhibition rates of tumor growth were compared between groups. The results showed that combined use of PJ34 and SAHA could synergistically inhibit the proliferation of liver cancer cell lines HepG2, Hep3B and HCC-LM3. The apoptosis rate of HepG2 cells treated with PJ34+SAHA was significantly higher than that of HepG2 cells treated with PJ34 or SAHA alone (P<0.05). In vivo, the tumor inhibition rates were 53.5%, 61.4% and 82.6% in PJ34, SAHA and PJ34+SAHA groups, respectively. The combined use of PJ34 and SAHA could significantly inhibit the xenograft tumor growth when compared with use of PJ34 or SAHA alone (P<0.05). It was led to conclude that PJ34 and SAHA can synergistically suppress the proliferation of liver cancer cells.
Collapse
Affiliation(s)
- Bin-Yong Liang
- Research Laboratory and Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Min Xiong
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gui-Bao Ji
- Research Laboratory and Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of General Surgery, Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Er-Lei Zhang
- Research Laboratory and Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zun-Yi Zhang
- Research Laboratory and Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ke-Shuai Dong
- Research Laboratory and Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Ping Chen
- Research Laboratory and Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhi-Yong Huang
- Research Laboratory and Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
44
|
Yan M, Shield-Artin K, Byrne D, Deb S, Waddell N, Haviv I, Fox SB. Comparative microRNA profiling of sporadic and BRCA1 associated basal-like breast cancers. BMC Cancer 2015; 15:506. [PMID: 26152113 PMCID: PMC4494690 DOI: 10.1186/s12885-015-1522-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 06/26/2015] [Indexed: 01/29/2023] Open
Abstract
Background While a number of studies have examined miRNA profiles across the molecular subtypes of breast cancer, it is unclear whether BRCA1 basal-like cancers have a specific miRNA profile. This study aims to compare grade independent miRNA expression in luminal cancers, sporadic and BRCA1 basal-type breast cancers. It also aims to ascertain an immunohistochemical profile regulated by BRCA1 specific miRNAs for potential diagnostic use. Methods miRNA expression was assessed in 11 BRCA1 basal, 16 sporadic basal, 17 luminal grade 3 cancers via microarrays. The expression of Cyclin D1, FOXP1, FIH-1, pan-ERβ, NRP1 and CD99, predicted to be regulated by BRCA1 specific miRNAs by computer prediction algorithms, was assessed via immunohistochemistry in a cohort of 35 BRCA1 and 52 sporadic basal-like cancers. Assessment of cyclin D1, FOXP1, NRP1 and CD99 expression was repeated on a validation cohort of 82 BRCA1 and 65 sporadic basal-like breast cancers. Results Unsupervised clustering of basal cancers resulted in a “sporadic” cluster of 11 cancers, and a “BRCA1” cluster of 16 cancers, including a subgroup composed entirely of 10 BRCA1 cancers. Compared with sporadic basal cancers, BRCA1 cancers showed reduced positivity for proteins predicted to be regulated by miRNAs: FOXP1 (6/20[30 %] vs. 37/49[76 %], p < 0.001), cyclin D1 (8/22[36 %] vs. 30/46[65 %], p = 0.025), NRP1 (2/20[10 %] vs. 23/46[50 %], p = 0.002). This was confirmed in the validation cohort (all p < 0.001). Negative staining for 2 or more out of FOXP1, cyclin D1 and NRP1 predicts germline BRCA1 mutation with a sensitivity of 92 %, specificity of 44 %, positive predictive value of 38 % and a negative predictive value of 94 %. Conclusion Sporadic and BRCA1 basal-like cancers have grade independent miRNA expression profiles. Furthermore miRNA driven differences in the expression of proteins in BRCA1 basal cancers may be detected via immunohistochemistry. These findings may have important diagnostic implications, as immunohistochemical assessment of basal cancers, in addition to the patient’s family and clinical history, may potentially identify patients who may benefit from BRCA1 gene testing. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1522-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Max Yan
- Department of Anatomical Pathology, Prince of Wales Hospital, School of Medical Sciences, University of New South Wales, Randwick, 2031, Australia.
| | | | - David Byrne
- Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, 3002, Australia.
| | - Siddhartha Deb
- Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, 3002, Australia.
| | - Nic Waddell
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, University of Queensland, St Lucia, Australia.
| | | | - Izhak Haviv
- Baker IDI Heart and Diabetes Institute, Prahran, 3004, Australia.
| | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, 3002, Australia.
| |
Collapse
|
45
|
Jones P, Wilcoxen K, Rowley M, Toniatti C. Niraparib: A Poly(ADP-ribose) Polymerase (PARP) Inhibitor for the Treatment of Tumors with Defective Homologous Recombination. J Med Chem 2015; 58:3302-14. [PMID: 25761096 DOI: 10.1021/jm5018237] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Poly(ADP-ribose) polymerases (PARPs) are involved in DNA repair following damage by endogenous or exogenous processes. It has become clear over the past decade that inhibition of PARP in the context of defects in other DNA repair mechanisms provide a tumor specific way to kill cancer cells. We describe the rationale for this approach and the design and discovery of niraparib, a potent PARP-1/2 inhibitor with good cell based activity, selectivity for cancer over normal cells, and oral bioavailability. Niraparib was characterized in a number of preclinical models before moving to phase I clinical trials, where it showed excellent human pharmacokinetics suitable for once a day oral dosing, achieved its pharmacodynamic target for PARP inhibition, and had promising activity in cancer patients. It is currently being tested in phase 3 clinical trials as maintenance therapy in ovarian cancer and as a treatment for breast cancer.
Collapse
Affiliation(s)
- Philip Jones
- †Istituto di Ricerche di Biologia Molecolare, Via Pontina km 30600, 00040 Pomezia, Italy
| | - Keith Wilcoxen
- ‡TESARO, Inc., 1000 Winter Street, Waltham, Massachusetts 02451, United States
| | - Michael Rowley
- †Istituto di Ricerche di Biologia Molecolare, Via Pontina km 30600, 00040 Pomezia, Italy
| | - Carlo Toniatti
- †Istituto di Ricerche di Biologia Molecolare, Via Pontina km 30600, 00040 Pomezia, Italy
| |
Collapse
|
46
|
Aleskandarany M, Caracappa D, Nolan CC, Macmillan RD, Ellis IO, Rakha EA, Green AR. DNA damage response markers are differentially expressed in BRCA-mutated breast cancers. Breast Cancer Res Treat 2015; 150:81-90. [PMID: 25690937 PMCID: PMC4344553 DOI: 10.1007/s10549-015-3306-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 02/09/2015] [Indexed: 01/07/2023]
Abstract
Cells have stringent DNA repair pathways that are specific for each different set of DNA lesions which is accomplished through the integration of complex array of proteins. However, BRCA-mutated breast cancer (BC) has defective DNA repair mechanisms. This study aims to investigate differential expression of a large panel of DNA repair markers to characterise DNA repair mechanisms in BRCA-associated tumours compared to sporadic tumours in an attempt to characterise these tumours in routine practice. Immunohistochemistry and tissue microarray technology were applied to a cohort of clinically annotated series of sporadic (n = 1849), BRCA1-mutated (n = 48), and BRCA2-mutated (n = 27) BC. The following DNA damage response (DDR) markers are used; BRCA1, BRCA2, RAD51, Ku70/Ku80, BARD, PARP1 (cleaved), PARP1 (non-cleaved), and P53 in addition to basal cytokeratins, ER, PR, and HER2. A significant proportion of BRCA1 tumours were positive for PARP1 (non-cleaved), and negative for BARD1 and RAD51 compared with sporadic BC. BRCA2 tumours were significantly positive for PARP1 (non-cleaved) compared with sporadic tumours. RAD51 was significantly higher in BRCA1 compared with BRCA2 tumours (p = 0.005). When BRCA1/2 BCs were compared to triple-negative (TN) sporadic tumours of the studied DDR proteins, BARD1 (p < 0.001), PARP1 (non-cleaved) (p < 0.001), and P53 (p = 0.002) remained significantly different in BRCA1/2 tumours compared with TN BC. DNA repair markers showed differential expression in BRCA-mutated tumours, with a substantial degree of disruption of DNA repair pathways in sporadic BC especially TN BC. DNA double-strand break (DSB) repair is assisted by PARP1 expression in BRCA-mutated tumours, whereas the loss of DSB repair via RAD51 is predominant in BRCA1 rather than BRCA2 BC.
Collapse
Affiliation(s)
- Mohammed Aleskandarany
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK,
| | | | | | | | | | | | | |
Collapse
|
47
|
Audeh MW. Novel treatment strategies in triple-negative breast cancer: specific role of poly(adenosine diphosphate-ribose) polymerase inhibition. Pharmgenomics Pers Med 2014; 7:307-16. [PMID: 25342917 PMCID: PMC4205934 DOI: 10.2147/pgpm.s39765] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Inhibitors of the poly(adenosine triphosphate-ribose) polymerase (PARP)-1 enzyme induce synthetic lethality in cancers with ineffective DNA (DNA) repair or homologous repair deficiency, and have shown promising clinical activity in cancers deficient in DNA repair due to germ-line mutation in BRCA1 and BRCA2. The majority of breast cancers arising in carriers of BRCA1 germ-line mutations, as well as half of those in BRCA2 carriers, are classified as triple-negative breast cancer (TNBC). TNBC is a biologically heterogeneous group of breast cancers characterized by the lack of immunohistochemical expression of the ER, PR, or HER2 proteins, and for which the current standard of care in systemic therapy is cytotoxic chemotherapy. Many "sporadic" cases of TNBC appear to have indicators of DNA repair dysfunction similar to those in BRCA-mutation carriers, suggesting the possible utility of PARP inhibitors in a subset of TNBC. Significant genetic heterogeneity has been observed within the TNBC cohort, creating challenges for interpretation of prior clinical trial data, and for the design of future clinical trials. Several PARP inhibitors are currently in clinical development in BRCA-mutated breast cancer. The use of PARP inhibitors in TNBC without BRCA mutation will require biomarkers that identify cancers with homologous repair deficiency in order to select patients likely to respond. Beyond mutations in the BRCA genes, dysfunction in other genes that interact with the homologous repair pathway may offer opportunities to induce synthetic lethality when combined with PARP inhibition.
Collapse
Affiliation(s)
- M William Audeh
- Division of Medical Oncology, Samuel Oschin Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
48
|
Norum JH, Andersen K, Sørlie T. Lessons learned from the intrinsic subtypes of breast cancer in the quest for precision therapy. Br J Surg 2014; 101:925-38. [PMID: 24849143 DOI: 10.1002/bjs.9562] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 04/16/2014] [Indexed: 01/06/2023]
Abstract
BACKGROUND Wide variability in breast cancer, between patients and within each individual neoplasm, adds confounding complexity to the treatment of the disease. In clinical practice, hormone receptor status has been used to classify breast tumours and to guide treatment. Modern classification systems should take the wide tumour heterogeneity into account to improve patient outcome. METHODS This article reviews the identification of the intrinsic molecular subtypes of breast cancer, their prognostic and therapeutic implications, and the impact of tumour heterogeneity on cancer progression and treatment. The possibility of functionally addressing tumour-specific characteristics in in vivo models to inform decisions for precision therapies is also discussed. RESULTS Despite the robust breast tumour classification system provided by gene expression profiling, heterogeneity is also evident within these molecular portraits. A complicating factor in breast cancer classification is the process of selective clonality within developing neoplasms. Phenotypically and functionally distinct clones representing the intratumour heterogeneity might confuse molecular classification. Molecular portraits of the heterogeneous primary tumour might not necessarily reflect the subclone of cancer cells that causes the disease to relapse. Studies of reciprocal relationships between cancer cell subpopulations within developing tumours are therefore needed, and are possible only in genetically engineered mouse models or patient-derived xenograft models, in which the treatment-induced selection pressure on individual cell clones can be mimicked. CONCLUSION In the future, more refined classifications, based on integration of information at several molecular levels, are required to improve treatment guidelines. Large-scale translational research efforts paved the way for identification of the intrinsic subtypes, and are still fundamental for ensuring future progress in cancer care.
Collapse
Affiliation(s)
- J H Norum
- Department of Genetics, Institute of Cancer Research, Oslo, Norway; Cancer Stem Cell Innovation Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway
| | | | | |
Collapse
|