51
|
Miao L, Yu Y, Ji Y, Zhang B, Yuan Z, Du Y, Zhu L, Wang R, Chen N, Yuan H. Association between BRCA1 P871L polymorphism and cancer risk: evidence from a meta-analysis. Oncotarget 2017; 8:30587-30594. [PMID: 28427168 PMCID: PMC5444767 DOI: 10.18632/oncotarget.15739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 02/13/2017] [Indexed: 01/23/2023] Open
Abstract
Breast cancer 1 (BRCA1) gene makes great contributions to the repair of DNA. The association between BRCA1 P871L polymorphism and cancer risk has been investigated in a growing number of studies, but the conclusions are not conclusive. To obtain a comprehensive conclusion, we performed a meta-analysis of 24 studies with 13762 cases and 22388 controls. The pooled results indicated that BRCA1 gene P871L variant decreased risk of overall cancer (homozygous model: odds ratio (OR) = 0.89, 95%confidence interval (CI) = 0.79-1.00; recessive model: OR = 0.89, 95% CI = 0.80-0.99). The stratified analysis observed decreased risk associated with BRCA1 P871L in subgroups among Asians and high score studies, but not Caucasians or low score studies. In conclusion, despite several limitations, this meta-analysis suggested that BRCA1 P871L genetic variation may be associated with decreased susceptibility to cancer.
Collapse
Affiliation(s)
- Limin Miao
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Yang Yu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Yefeng Ji
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Bo Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Zhiyao Yuan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Yifei Du
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Longbiao Zhu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Ruixia Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Ning Chen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Hua Yuan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
52
|
Bower JJ, Vance LD, Psioda M, Smith-Roe SL, Simpson DA, Ibrahim JG, Hoadley KA, Perou CM, Kaufmann WK. Patterns of cell cycle checkpoint deregulation associated with intrinsic molecular subtypes of human breast cancer cells. NPJ Breast Cancer 2017; 3:9. [PMID: 28649649 PMCID: PMC5445620 DOI: 10.1038/s41523-017-0009-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 02/07/2017] [Indexed: 12/31/2022] Open
Abstract
Genomic instability is a hallmark of breast cancer, contributes to tumor heterogeneity, and influences chemotherapy resistance. Although Gap 2 and mitotic checkpoints are thought to prevent genomic instability, the role of these checkpoints in breast cancer is poorly understood. Here, we assess the Gap 2 and mitotic checkpoint functions of 24 breast cancer and immortalized mammary epithelial cell lines representing four of the six intrinsic molecular subtypes of breast cancer. We found that patterns of cell cycle checkpoint deregulation were associated with the intrinsic molecular subtype of breast cancer cell lines. Specifically, the luminal B and basal-like cell lines harbored two molecularly distinct Gap 2/mitosis checkpoint defects (impairment of the decatenation Gap 2 checkpoint and the spindle assembly checkpoint, respectively). All subtypes of breast cancer cell lines examined displayed aberrant DNA synthesis/Gap 2/mitosis progression and the basal-like and claudin-low cell lines exhibited increased percentages of chromatid cohesion defects. Furthermore, a decatenation Gap 2 checkpoint gene expression signature identified in the cell line panel correlated with clinical outcomes in breast cancer patients, suggesting that breast tumors may also harbor defects in decatenation Gap 2 checkpoint function. Taken together, these data imply that pharmacological targeting of signaling pathways driving these phenotypes may lead to the development of novel personalized treatment strategies for the latter two subtypes which currently lack targeted therapeutic options because of their triple negative breast cancer status.
Collapse
Affiliation(s)
- Jacquelyn J. Bower
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Leah D. Vance
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Matthew Psioda
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Stephanie L. Smith-Roe
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, National Institutes of Health, Research Triangle Park, NC 27709 USA
| | - Dennis A. Simpson
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Joseph G. Ibrahim
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Katherine A. Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Charles M. Perou
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - William K. Kaufmann
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| |
Collapse
|
53
|
Anders CK, Abramson V, Tan T, Dent R. The Evolution of Triple-Negative Breast Cancer: From Biology to Novel Therapeutics. Am Soc Clin Oncol Educ Book 2017; 35:34-42. [PMID: 27249684 DOI: 10.1200/edbk_159135] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Triple-negative breast cancer (TNBC) is clinically defined as lacking expression of the estrogen receptor (ER), progesterone receptor (ER), and HER2. Historically, TNBC has been characterized by an aggressive natural history and worse disease-specific outcomes compared with other breast cancer subtypes. The advent of next-generation sequencing (NGS) has allowed for the dissection of TNBC into molecular subtypes (i.e., basal-like, claudin-low). Within TNBC, several subtypes have emerged as "immune-activated," consistently illustrating better disease outcome. In addition, NGS has revealed a host of molecular features characteristic of TNBC, including high rates of TP53 mutations, PI3K and MEK pathway activation, and genetic similarities to serous ovarian cancers, including inactivation of the BRCA pathway. Identified genetic vulnerabilities of TNBC have led to promising therapeutic approaches, including DNA-damaging agents (i.e., platinum salts and PARP inhibitors), as well as immunotherapy. Platinum salts are routinely incorporated into the treatment of metastatic TNBC; however, best outcomes are observed among those with deficiencies in the BRCA pathway. Although the incorporation of platinum in the neoadjuvant care of patients with TNBC yields higher pathologic complete response (pCR) rates, the impact on longer-term outcome is less clear. The presence of immune infiltrate in TNBC has shown both a predictive and prognostic role. Checkpoint inhibitors, including PD-1 and PD-L1 inhibitors, are under investigation in the setting of metastatic TNBC and have shown responses in initial clinical trials. Finally, matching emerging therapeutic strategies to optimal subtype of TNBC is of utmost importance as we design future research strategies to improve patient outcome.
Collapse
Affiliation(s)
- Carey K Anders
- From the Department of Medicine, Vanderbilt University, Vanderbilt-Ingram Cancer Center, Nashville, TN; Department of Medicine, National Cancer Center Singapore, Singapore; Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC; UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | - Vandana Abramson
- From the Department of Medicine, Vanderbilt University, Vanderbilt-Ingram Cancer Center, Nashville, TN; Department of Medicine, National Cancer Center Singapore, Singapore; Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC; UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | - Tira Tan
- From the Department of Medicine, Vanderbilt University, Vanderbilt-Ingram Cancer Center, Nashville, TN; Department of Medicine, National Cancer Center Singapore, Singapore; Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC; UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | - Rebecca Dent
- From the Department of Medicine, Vanderbilt University, Vanderbilt-Ingram Cancer Center, Nashville, TN; Department of Medicine, National Cancer Center Singapore, Singapore; Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC; UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| |
Collapse
|
54
|
Yao H, He G, Yan S, Chen C, Song L, Rosol TJ, Deng X. Triple-negative breast cancer: is there a treatment on the horizon? Oncotarget 2017; 8:1913-1924. [PMID: 27765921 PMCID: PMC5352107 DOI: 10.18632/oncotarget.12284] [Citation(s) in RCA: 268] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 09/17/2016] [Indexed: 12/28/2022] Open
Abstract
Triple-negative breast cancer (TNBC), which accounts for 15-20% of all breast cancers, does not express estrogen receptor (ER) or progesterone receptor (PR) and lacks human epidermal growth factor receptor 2 (HER2) overexpression or amplification. These tumors have a more aggressive phenotype and a poorer prognosis due to the high propensity for metastatic progression and absence of specific targeted treatments. Patients with TNBC do not benefit from hormonal or trastuzumab-based targeted therapies because of the loss of target receptors. Although these patients respond to chemotherapeutic agents such as taxanes and anthracyclines better than other subtypes of breast cancer, prognosis remains poor. A group of targeted therapies under investigation showed favorable results in TNBC, especially in cancers with BRCA mutation. The lipid-lowering statins (3-hydroxy-3-methyl-glutaryl coenzyme A reductase inhibitors), including lovastatin and simvastatin, have been shown to preferentially target TNBC compared with non-TNBC. These statins hold great promise for the management of TNBC. Only with the understanding of the molecular basis for the preference of statins for TNBC and more investigations in clinical trials can they be reformulated into a clinically approved drug against TNBC.
Collapse
Affiliation(s)
- Hui Yao
- Department of Pathology, Hunan Normal University Medical College, Changsha, Hunan, China
| | - Guangchun He
- Department of Pathology, Hunan Normal University Medical College, Changsha, Hunan, China
| | - Shichao Yan
- Department of Pathology, Hunan Normal University Medical College, Changsha, Hunan, China
| | - Chao Chen
- Department of Pathology, Hunan Normal University Medical College, Changsha, Hunan, China
| | - Liujiang Song
- Department of Pediatrics, Hunan Normal University Medical College, Changsha, Hunan, China
| | - Thomas J. Rosol
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Xiyun Deng
- Department of Pathology, Hunan Normal University Medical College, Changsha, Hunan, China
| |
Collapse
|
55
|
Kirova YM, Fourquet A. Concurrent Use of Radiation Therapy and Targeted Molecules in the Breast Cancer Treatment. Breast Cancer 2017. [DOI: 10.1007/978-3-319-48848-6_54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
56
|
Dréan A, Lord CJ, Ashworth A. PARP inhibitor combination therapy. Crit Rev Oncol Hematol 2016; 108:73-85. [PMID: 27931843 DOI: 10.1016/j.critrevonc.2016.10.010] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 09/02/2016] [Accepted: 10/26/2016] [Indexed: 01/02/2023] Open
Abstract
In 2014, olaparib (Lynparza) became the first PARP (Poly(ADP-ribose) polymerase) inhibitor to be approved for the treatment of cancer. When used as single agents, PARP inhibitors can selectively target tumour cells with BRCA1 or BRCA2 tumour suppressor gene mutations through synthetic lethality. However, PARP inhibition also shows considerable promise when used together with other therapeutic agents. Here, we summarise both the pre-clinical and clinical evidence for the utility of such combinations and discuss the future prospects and challenges for PARP inhibitor combinatorial therapies.
Collapse
Affiliation(s)
- Amy Dréan
- The CRUK Gene Function Laboratory, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Christopher J Lord
- The CRUK Gene Function Laboratory, The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK.
| | - Alan Ashworth
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
57
|
Telli ML, Timms KM, Reid J, Hennessy B, Mills GB, Jensen KC, Szallasi Z, Barry WT, Winer EP, Tung NM, Isakoff SJ, Ryan PD, Greene-Colozzi A, Gutin A, Sangale Z, Iliev D, Neff C, Abkevich V, Jones JT, Lanchbury JS, Hartman AR, Garber JE, Ford JM, Silver DP, Richardson AL. Homologous Recombination Deficiency (HRD) Score Predicts Response to Platinum-Containing Neoadjuvant Chemotherapy in Patients with Triple-Negative Breast Cancer. Clin Cancer Res 2016. [PMID: 26957554 DOI: 10.1158/1078-0432.ccr-15-2477] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE BRCA1/2-mutated and some sporadic triple-negative breast cancers (TNBC) have DNA repair defects and are sensitive to DNA-damaging therapeutics. Recently, three independent DNA-based measures of genomic instability were developed on the basis of loss of heterozygosity (LOH), telomeric allelic imbalance (TAI), and large-scale state transitions (LST). EXPERIMENTAL DESIGN We assessed a combined homologous recombination deficiency (HRD) score, an unweighted sum of LOH, TAI, and LST scores, in three neoadjuvant TNBC trials of platinum-containing therapy. We then tested the association of HR deficiency, defined as HRD score ≥42 or BRCA1/2 mutation, with response to platinum-based therapy. RESULTS In a trial of neoadjuvant platinum, gemcitabine, and iniparib, HR deficiency predicted residual cancer burden score of 0 or I (RCB 0/I) and pathologic complete response (pCR; OR = 4.96, P = 0.0036; OR = 6.52, P = 0.0058). HR deficiency remained a significant predictor of RCB 0/I when adjusted for clinical variables (OR = 5.86, P = 0.012). In two other trials of neoadjuvant cisplatin therapy, HR deficiency predicted RCB 0/I and pCR (OR = 10.18, P = 0.0011; OR = 17.00, P = 0.0066). In a multivariable model of RCB 0/I, HR deficiency retained significance when clinical variables were included (OR = 12.08, P = 0.0017). When restricted to BRCA1/2 nonmutated tumors, response was higher in patients with high HRD scores: RCB 0/I P = 0.062, pCR P = 0.063 in the neoadjuvant platinum, gemcitabine, and iniparib trial; RCB 0/I P = 0.0039, pCR P = 0.018 in the neoadjuvant cisplatin trials. CONCLUSIONS HR deficiency identifies TNBC tumors, including BRCA1/2 nonmutated tumors more likely to respond to platinum-containing therapy. Clin Cancer Res; 22(15); 3764-73. ©2016 AACR.
Collapse
Affiliation(s)
- Melinda L Telli
- Stanford University School of Medicine, Stanford, California
| | | | - Julia Reid
- Myriad Genetics Inc., Salt Lake City, Utah
| | - Bryan Hennessy
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gordon B Mills
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Zoltan Szallasi
- Denmark Technical University, Lyngby, Denmark. Children's Hospital Boston, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - William T Barry
- Harvard Medical School, Boston, Massachusetts. Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Eric P Winer
- Harvard Medical School, Boston, Massachusetts. Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nadine M Tung
- Harvard Medical School, Boston, Massachusetts. Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Steven J Isakoff
- Harvard Medical School, Boston, Massachusetts. Massachusetts General Hospital, Boston, Massachusetts
| | - Paula D Ryan
- Massachusetts General Hospital, Boston, Massachusetts
| | | | | | | | | | - Chris Neff
- Myriad Genetics Inc., Salt Lake City, Utah
| | | | | | | | | | - Judy E Garber
- Harvard Medical School, Boston, Massachusetts. Dana-Farber Cancer Institute, Boston, Massachusetts
| | - James M Ford
- Stanford University School of Medicine, Stanford, California
| | - Daniel P Silver
- Harvard Medical School, Boston, Massachusetts. Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Andrea L Richardson
- Harvard Medical School, Boston, Massachusetts. Dana-Farber Cancer Institute, Boston, Massachusetts. Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
58
|
Telli ML, Timms KM, Reid J, Hennessy B, Mills GB, Jensen KC, Szallasi Z, Barry WT, Winer EP, Tung NM, Isakoff SJ, Ryan PD, Greene-Colozzi A, Gutin A, Sangale Z, Iliev D, Neff C, Abkevich V, Jones JT, Lanchbury JS, Hartman AR, Garber JE, Ford JM, Silver DP, Richardson AL. Homologous Recombination Deficiency (HRD) Score Predicts Response to Platinum-Containing Neoadjuvant Chemotherapy in Patients with Triple-Negative Breast Cancer. Clin Cancer Res 2016; 22:3764-73. [PMID: 26957554 DOI: 10.1158/1078-0432.ccr-15-2477] [Citation(s) in RCA: 760] [Impact Index Per Article: 84.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/09/2016] [Indexed: 12/15/2022]
Abstract
PURPOSE BRCA1/2-mutated and some sporadic triple-negative breast cancers (TNBC) have DNA repair defects and are sensitive to DNA-damaging therapeutics. Recently, three independent DNA-based measures of genomic instability were developed on the basis of loss of heterozygosity (LOH), telomeric allelic imbalance (TAI), and large-scale state transitions (LST). EXPERIMENTAL DESIGN We assessed a combined homologous recombination deficiency (HRD) score, an unweighted sum of LOH, TAI, and LST scores, in three neoadjuvant TNBC trials of platinum-containing therapy. We then tested the association of HR deficiency, defined as HRD score ≥42 or BRCA1/2 mutation, with response to platinum-based therapy. RESULTS In a trial of neoadjuvant platinum, gemcitabine, and iniparib, HR deficiency predicted residual cancer burden score of 0 or I (RCB 0/I) and pathologic complete response (pCR; OR = 4.96, P = 0.0036; OR = 6.52, P = 0.0058). HR deficiency remained a significant predictor of RCB 0/I when adjusted for clinical variables (OR = 5.86, P = 0.012). In two other trials of neoadjuvant cisplatin therapy, HR deficiency predicted RCB 0/I and pCR (OR = 10.18, P = 0.0011; OR = 17.00, P = 0.0066). In a multivariable model of RCB 0/I, HR deficiency retained significance when clinical variables were included (OR = 12.08, P = 0.0017). When restricted to BRCA1/2 nonmutated tumors, response was higher in patients with high HRD scores: RCB 0/I P = 0.062, pCR P = 0.063 in the neoadjuvant platinum, gemcitabine, and iniparib trial; RCB 0/I P = 0.0039, pCR P = 0.018 in the neoadjuvant cisplatin trials. CONCLUSIONS HR deficiency identifies TNBC tumors, including BRCA1/2 nonmutated tumors more likely to respond to platinum-containing therapy. Clin Cancer Res; 22(15); 3764-73. ©2016 AACR.
Collapse
Affiliation(s)
- Melinda L Telli
- Stanford University School of Medicine, Stanford, California
| | | | - Julia Reid
- Myriad Genetics Inc., Salt Lake City, Utah
| | - Bryan Hennessy
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gordon B Mills
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Zoltan Szallasi
- Denmark Technical University, Lyngby, Denmark. Children's Hospital Boston, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - William T Barry
- Harvard Medical School, Boston, Massachusetts. Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Eric P Winer
- Harvard Medical School, Boston, Massachusetts. Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nadine M Tung
- Harvard Medical School, Boston, Massachusetts. Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Steven J Isakoff
- Harvard Medical School, Boston, Massachusetts. Massachusetts General Hospital, Boston, Massachusetts
| | - Paula D Ryan
- Massachusetts General Hospital, Boston, Massachusetts
| | | | | | | | | | - Chris Neff
- Myriad Genetics Inc., Salt Lake City, Utah
| | | | | | | | | | - Judy E Garber
- Harvard Medical School, Boston, Massachusetts. Dana-Farber Cancer Institute, Boston, Massachusetts
| | - James M Ford
- Stanford University School of Medicine, Stanford, California
| | - Daniel P Silver
- Harvard Medical School, Boston, Massachusetts. Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Andrea L Richardson
- Harvard Medical School, Boston, Massachusetts. Dana-Farber Cancer Institute, Boston, Massachusetts. Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
59
|
Synergetic Effects of PARP Inhibitor AZD2281 and Cisplatin in Oral Squamous Cell Carcinoma in Vitro and in Vivo. Int J Mol Sci 2016; 17:272. [PMID: 26927065 PMCID: PMC4813136 DOI: 10.3390/ijms17030272] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/08/2016] [Accepted: 02/17/2016] [Indexed: 01/03/2023] Open
Abstract
Cisplatin is a commonly used chemotherapeutic drug for treatment of oral carcinoma, and combinatorial effects are expected to exert greater therapeutic efficacy compared with monotherapy. Poly(ADP-ribosyl)ation is reported to be involved in a variety of cellular processes, such as DNA repair, cell death, telomere regulation, and genomic stability. Based on these properties, poly(ADP-ribose) polymerase (PARP) inhibitors are used for treatment of cancers, such as BRCA1/2 mutated breast and ovarian cancers, or certain solid cancers in combination with anti-cancer drugs. However, the effects on oral cancer have not been fully evaluated. In this study, we examined the effects of PARP inhibitor on the survival of human oral cancer cells in vitro and xenografted tumors derived from human oral cancer cells in vivo. In vitro effects were assessed by microculture tetrazolium and survival assays. The PARP inhibitor AZD2281 (olaparib) showed synergetic effects with cisplatin in a dose-dependent manner. Combinatorial treatment with cisplatin and AZD2281 significantly inhibited xenografted tumor growth compared with single treatment of cisplatin or AZD2281. Histopathological analysis revealed that cisplatin and AZD2281 increased TUNEL-positive cells and decreased Ki67- and CD31-positive cells. These results suggest that PARP inhibitors have the potential to improve therapeutic strategies for oral cancer.
Collapse
|
60
|
Su S, Ding Y, Li Y, Wu Y, Nie G. Integration of photothermal therapy and synergistic chemotherapy by a porphyrin self-assembled micelle confers chemosensitivity in triple-negative breast cancer. Biomaterials 2016; 80:169-178. [DOI: 10.1016/j.biomaterials.2015.11.058] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/16/2015] [Accepted: 11/29/2015] [Indexed: 12/19/2022]
|
61
|
Ávila-Arroyo S, Nuñez GS, García-Fernández LF, Galmarini CM. Synergistic Effect of Trabectedin and Olaparib Combination Regimen in Breast Cancer Cell Lines. J Breast Cancer 2015; 18:329-38. [PMID: 26770239 PMCID: PMC4705084 DOI: 10.4048/jbc.2015.18.4.329] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 10/06/2015] [Indexed: 01/01/2023] Open
Abstract
Purpose Trabectedin induces synthetic lethality in tumor cells carrying defects in homologous recombinant DNA repair. We evaluated the effect of concomitant inhibition of nucleotide-excision repair and poly (ADP-ribose) polymerase (PARP) activity with trabectedin and PARP inhibitors, respectively, and whether the synthetic lethality effect had the potential for a synergistic effect in breast cancer cell lines. Additionally, we investigated if this approach remained effective in BRCA1-positive breast tumor cells. Methods We have evaluated the in vitro synergistic effect of combinations of trabectedin and three different PARP inhibitors (veliparib, olaparib, and iniparib) in four breast cancer cell lines, each presenting a different BRCA1 genetic background. Antiproliferative activity, DNA damage, cell cycle perturbations and poly(ADP-ribosyl)ation were assessed by MTT assay, comet assay, flow cytometry and western blot, respectively. Results The combination of trabectedin and olaparib was synergistic in all the breast cancer cell lines tested. Our data indicated that the synergy persisted regardless of the BRCA1 status of the tumor cells. Combination treatment was associated with a strong accumulation of double-stranded DNA breaks, G2/M arrest, and apoptotic cell death. Synergistic effects were not observed when trabectedin was combined with veliparib or iniparib. Conclusion Collectively, our results indicate that the combination of trabectedin and olaparib induces an artificial synthetic lethality effect that can be used to kill breast cancer cells, independent of BRCA1 status.
Collapse
Affiliation(s)
- Sonia Ávila-Arroyo
- Cell Biology and Pharmacogenomics Department, PharmaMar S.A., Madrid, Spain
| | | | | | - Carlos M Galmarini
- Cell Biology and Pharmacogenomics Department, PharmaMar S.A., Madrid, Spain
| |
Collapse
|
62
|
Yadav BS, Chanana P, Jhamb S. Biomarkers in triple negative breast cancer: A review. World J Clin Oncol 2015; 6:252-263. [PMID: 26677438 PMCID: PMC4675910 DOI: 10.5306/wjco.v6.i6.252] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 09/03/2015] [Accepted: 10/01/2015] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is an intrinsically heterogeneous disease. In the world about 1 million cases of breast cancer are diagnosed annually and more than 170000 are triple-negative. Characteristic feature of triple negative breast cancer (TNBC) is that it lacks expression of oestrogen, progesterone and human epidermal growth factor receptor-2/neu receptors. They comprise 15%-20% of all breast cancers. We did a systematic review of PubMed and conference databases to identify studies published on biomarkers in TNBC. We included studies with biomarkers including: Epidermal growth factor receptor, vascular endothelial growth factor, c-Myc, C-kit and basal cytokeratins, Poly(ADP-ribose) polymerase-1, p53, tyrosinase kinases, m-TOR, heat and shock proteins and TOP-2A in TNBC. We also looked for studies published on synthetic lethality and inhibition of angiogenesis, growth, and survival pathways. TNBC is a complex disease subtype with many subclasses. Majority TNBC have a basal-like molecular phenotype by gene expression profiling. Their clinical and pathologic features overlap with hereditary BRCA1 related breast cancers. Management of these tumours is a challenge to the clinician because of its aggressive behaviour, poor outcome, and absence of targeted therapies. As the complexity of this disease is being simplified over time new targets are also being discovered for the treatment of this disease. There are many biomarkers in TNBC being used in clinical practice. Biomarkers may be useful as prognostic or predictive indicators as well as suggest possible targets for novel therapies. Many targeted agents are being studied for treatment of TNBC.
Collapse
|
63
|
Abstract
Recent discoveries both in cell proliferation and survival mechanisms and new antineoplastic agents have led to deep change in the breast cancer treatment paradigm. Nonetheless, all of the progress in knowledge and strategy has not been enough to overcome mechanisms of escape and resistance put in place by the tumor cells. New targeted agents mean new possibilities for combinations, a viable option to try to stop compensatory pathways of tumor growth activated in response to therapeutics. The main challenges in designing a combined therapy come from the variety of subtypes of breast cancer (luminal A, luminal B, HER2-enriched, and basal-like) and from the multitude of pathways each subtype can exploit. Recent research has focused on dual blockade of HER2 (trastuzumab-lapatinib; trastuzumab-pertuzumab) and concomitant blockade of the endocrine driver and other pathways such as the PI3K/AKT/mTOR pathway (everolimus-exemestane), HER2 (trastuzumab/lapatinib-endocrine therapy) and the cell cycle through cyclin-dependent kinase inhibition (letrozole-palbociclib). This combined and personalized approach to treatment needs a profound knowledge of the mechanisms leading to proliferation in each tumor subtype. Deepening our understanding of tumor growth is mandatory to keep improving the efficacy of combination therapy.
Collapse
Affiliation(s)
- Elisa Zanardi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Giacomo Bregni
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | |
Collapse
|
64
|
|
65
|
Histone deacetylase inhibitor treatment induces 'BRCAness' and synergistic lethality with PARP inhibitor and cisplatin against human triple negative breast cancer cells. Oncotarget 2015; 5:5637-50. [PMID: 25026298 PMCID: PMC4170637 DOI: 10.18632/oncotarget.2154] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
There is an unmet need to develop new, more effective and safe therapies for the aggressive forms of triple negative breast cancers (TNBCs). While up to 20% of women under 50 years of age with TNBC harbor germline mutations in BRCA1, and these tumors are sensitive to treatment with poly(ADP) ribose polymerase inhibitors, a majority of TNBCs lack BRCA1 mutations or loss of expression. Findings presented here demonstrate that by attenuating the levels of DNA damage response and homologous recombination proteins, pan-histone deacetylase inhibitor (HDI) treatment induces ‘BRCAness’ and sensitizes TNBC cells lacking BRCA1 to lethal effects of PARP inhibitor or cisplatin. Treatment with HDI also induced hyperacetylation of nuclear hsp90. Similar effects were observed following shRNA-mediated depletion of HDAC3, confirming its role as the deacetylase for nuclear HSP90. Furthermore, cotreatment with HDI and ABT-888 induced significantly more DNA strand breaks than either agent alone, and synergistically induced apoptosis of TNBC cells. Notably, co-treatment with HDI and ABT-888 significantly reduced in vivo tumor growth and markedly improved the survival of mice bearing TNBC cell xenografts. These findings support the rationale to interrogate the clinical activity of this novel combination against human TNBC, irrespective of its expression of mutant BRCA1.
Collapse
|
66
|
Telli ML, Jensen KC, Vinayak S, Kurian AW, Lipson JA, Flaherty PJ, Timms K, Abkevich V, Schackmann EA, Wapnir IL, Carlson RW, Chang PJ, Sparano JA, Head B, Goldstein LJ, Haley B, Dakhil SR, Reid JE, Hartman AR, Manola J, Ford JM. Phase II Study of Gemcitabine, Carboplatin, and Iniparib As Neoadjuvant Therapy for Triple-Negative and BRCA1/2 Mutation-Associated Breast Cancer With Assessment of a Tumor-Based Measure of Genomic Instability: PrECOG 0105. J Clin Oncol 2015; 33:1895-901. [PMID: 25847929 DOI: 10.1200/jco.2014.57.0085] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
PURPOSE This study was designed to assess efficacy, safety, and predictors of response to iniparib in combination with gemcitabine and carboplatin in early-stage triple-negative and BRCA1/2 mutation-associated breast cancer. PATIENTS AND METHODS This single-arm phase II study enrolled patients with stage I to IIIA (T ≥ 1 cm) estrogen receptor-negative (≤ 5%), progesterone receptor-negative (≤ 5%), and human epidermal growth factor receptor 2-negative or BRCA1/2 mutation-associated breast cancer. Neoadjuvant gemcitabine (1,000 mg/m(2) intravenously [IV] on days 1 and 8), carboplatin (area under curve of 2 IV on days 1 and 8), and iniparib (5.6 mg/kg IV on days 1, 4, 8, and 11) were administered every 21 days for four cycles, until the protocol was amended to six cycles. The primary end point was pathologic complete response (no invasive carcinoma in breast or axilla). All patients underwent comprehensive BRCA1/2 genotyping, and homologous recombination deficiency was assessed by loss of heterozygosity (HRD-LOH) in pretreatment core breast biopsies. RESULTS Among 80 patients, median age was 48 years; 19 patients (24%) had germline BRCA1 or BRCA2 mutations; clinical stage was I (13%), IIA (36%), IIB (36%), and IIIA (15%). Overall pathologic complete response rate in the intent-to-treat population (n = 80) was 36% (90% CI, 27 to 46). Mean HRD-LOH scores were higher in responders compared with nonresponders (P = .02) and remained significant when BRCA1/2 germline mutations carriers were excluded (P = .021). CONCLUSION Preoperative combination of gemcitabine, carboplatin, and iniparib is active in the treatment of early-stage triple-negative and BRCA1/2 mutation-associated breast cancer. The HRD-LOH assay was able to identify patients with sporadic triple-negative breast cancer lacking a BRCA1/2 mutation, but with an elevated HRD-LOH score, who achieved a favorable pathologic response. Confirmatory controlled trials are warranted.
Collapse
Affiliation(s)
- Melinda L Telli
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA.
| | - Kristin C Jensen
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Shaveta Vinayak
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Allison W Kurian
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Jafi A Lipson
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Patrick J Flaherty
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Kirsten Timms
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Victor Abkevich
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Elizabeth A Schackmann
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Irene L Wapnir
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Robert W Carlson
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Pei-Jen Chang
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Joseph A Sparano
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Bobbie Head
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Lori J Goldstein
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Barbara Haley
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Shaker R Dakhil
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Julia E Reid
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Anne-Renee Hartman
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - Judith Manola
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| | - James M Ford
- Melinda L. Telli, Kristin C. Jensen, Shaveta Vinayak, Allison W. Kurian, Jafi A. Lipson, Patrick J. Flaherty, Elizabeth A. Schackmann, Irene L. Wapnir, Robert W. Carlson, Pei-Jen Chang, and James M. Ford, Stanford University School of Medicine, Stanford; Bobbie Head, Marin Specialty Care, Greenbrae, CA; Kirsten Timms, Victor Abkevich, Julia E. Reid, and Anne-Renee Hartman, Myriad Genetics, Salt Lake City, UT; Joseph A. Sparano, Albert Einstein College of Medicine, New York, NY; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia, PA; Barbara Haley, University of Texas Southwestern Medical Center, Dallas, TX; Shaker R. Dakhil, Cancer Center of Kansas, Wichita, KS; and Judith Manola, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
67
|
Polotskaia A, Xiao G, Reynoso K, Martin C, Qiu WG, Hendrickson RC, Bargonetti J. Proteome-wide analysis of mutant p53 targets in breast cancer identifies new levels of gain-of-function that influence PARP, PCNA, and MCM4. Proc Natl Acad Sci U S A 2015; 112:E1220-9. [PMID: 25733866 PMCID: PMC4371979 DOI: 10.1073/pnas.1416318112] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The gain-of-function mutant p53 (mtp53) transcriptome has been studied, but, to date, no detailed analysis of the mtp53-associated proteome has been described. We coupled cell fractionation with stable isotope labeling with amino acids in cell culture (SILAC) and inducible knockdown of endogenous mtp53 to determine the mtp53-driven proteome. Our fractionation data highlight the underappreciated biology that missense mtp53 proteins R273H, R280K, and L194F are tightly associated with chromatin. Using SILAC coupled to tandem MS, we identified that R273H mtp53 expression in MDA-MB-468 breast cancer cells up- and down-regulated multiple proteins and metabolic pathways. Here we provide the data set obtained from sequencing 73,154 peptide pairs that then corresponded to 3,010 proteins detected under reciprocal labeling conditions. Importantly, the high impact regulated targets included the previously identified transcriptionally regulated mevalonate pathway proteins but also identified two new levels of mtp53 protein regulation for nontranscriptional targets. Interestingly, mtp53 depletion profoundly influenced poly(ADP ribose) polymerase 1 (PARP1) localization, with increased cytoplasmic and decreased chromatin-associated protein. An enzymatic PARP shift occurred with high mtp53 expression, resulting in increased poly-ADP-ribosylated proteins in the nucleus. Mtp53 increased the level of proliferating cell nuclear antigen (PCNA) and minichromosome maintenance 4 (MCM4) proteins without changing the amount of pcna and mcm4 transcripts. Pathway enrichment analysis ranked the DNA replication pathway above the cholesterol biosynthesis pathway as a R273H mtp53 activated proteomic target. Knowledge of the proteome diversity driven by mtp53 suggests that DNA replication and repair pathways are major targets of mtp53 and highlights consideration of combination chemotherapeutic strategies targeting cholesterol biosynthesis and PARP inhibition.
Collapse
Affiliation(s)
- Alla Polotskaia
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065; and
| | - Gu Xiao
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065; and
| | - Katherine Reynoso
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065; and
| | - Che Martin
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065; and
| | - Wei-Gang Qiu
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065; and
| | - Ronald C Hendrickson
- Proteomics Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Jill Bargonetti
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065; and
| |
Collapse
|
68
|
Alluri P, Newman LA. Basal-like and triple-negative breast cancers: searching for positives among many negatives. Surg Oncol Clin N Am 2015; 23:567-77. [PMID: 24882351 DOI: 10.1016/j.soc.2014.03.003] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Triple-negative breast cancers (TNBC) are defined by their failure to express the estrogen receptor, progesterone receptor, and HER2/neu protein markers. This basic feature is clinically relevant because it indicates that these cancers cannot be managed with endocrine or anti-HER2 systemic therapies. Furthermore, most TNBC cases are also characterized as being of the genetically defined basal subtype, which is an inherently and biologically more aggressive pattern of disease. The two terms, however, are not synonymous, and some TNBC cases are prognostically more favorable. TNBC differs from non-TNBC in risk-factor profile, pattern, and rate of metastatic spread.
Collapse
Affiliation(s)
- Prasanna Alluri
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Lisa A Newman
- Breast Care Center, University of Michigan Comprehensive Cancer Center, 1500 East Medical Center Drive, Ann Arbor, MI 48167, USA.
| |
Collapse
|
69
|
Montariello D, Troiano A, Di Girolamo D, Beneke S, Calabrò V, Quesada P. Effect of poly(ADP-ribose)polymerase and DNA topoisomerase I inhibitors on the p53/p63-dependent survival of carcinoma cells. Biochem Pharmacol 2015; 94:212-9. [PMID: 25667043 DOI: 10.1016/j.bcp.2015.01.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 01/20/2015] [Accepted: 01/21/2015] [Indexed: 11/30/2022]
Abstract
Depending on their genetic background (p53(wt) versus p53(null)), carcinoma cells are more or less sensitive to drug-induced cell cycle arrest and/or apoptosis. Among the members of the p53 family, p63 is characterized by two N-terminal isoforms, TAp63 and ΔNp63. TAp63 isoform has p53-like functions, while ΔNp63 acts as a dominant negative inhibitor of p53. We have previously published that TAp63 is involved in poly(ADP-ribose)polymerase-1 (PARP-1) signaling of DNA damage deriving from DNA topoisomerase I (TOP I) inhibition in carcinoma cells. In the present study, we treated MCF7 breast carcinoma cells (p53(+)/ΔNp63(-)) or SCC022 (p53(-)/ΔNp63(+)) squamous carcinoma cells with the TOP I inhibitor topotecan (TPT) and the PJ34 PARP inhibitor, to compare their effects in the two different cell contexts. In MCF7 cells, we found that PJ34 addition reverts TPT-dependent PARP-1 auto-modification and triggers caspase-dependent PARP-1 proteolysis. Moreover, TPT as single agent stimulates p53(ser15) phosphorylation, p53 PARylation and occupancy of the p21WAF promoter by p53 resulting in an increase of p21WAF expression. Interestingly, PJ34 in combination with TPT enhances p53 occupancy at the BAX promoter and is associated with increased BAX protein level. In SCC022 cells, instead, TPT+PJ34 combined treatment reduces the level of the anti-apoptotic ΔNp63α protein without inducing apoptosis. Remarkably, in such cells, either exogenous p53 or TAp63 can rescue the apoptotic program in response to the treatment. All together our results suggest that in cancer cells PARP inhibitor(s) can operate in the choice between growth arrest and apoptosis by modulating p53 family-dependent signal.
Collapse
Affiliation(s)
| | - Annaelena Troiano
- Department of Biology, University of Naples "Federico II", Naples, Italy
| | | | - Sascha Beneke
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Viola Calabrò
- Department of Biology, University of Naples "Federico II", Naples, Italy
| | - Piera Quesada
- Department of Biology, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
70
|
Hussain A, Mohsin J, Prabhu SA, Begum S, Nusri QEA, Harish G, Javed E, Khan MA, Sharma C. Sulforaphane inhibits growth of human breast cancer cells and augments the therapeutic index of the chemotherapeutic drug, gemcitabine. Asian Pac J Cancer Prev 2015; 14:5855-60. [PMID: 24289589 DOI: 10.7314/apjcp.2013.14.10.5855] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Phytochemicals are among the natural chemopreventive agents with most potential for delaying, blocking or reversing the initiation and promotional events of carcinogenesis. They therefore offer cancer treatment strategies to reduce cancer related death. One such promising chemopreventive agent which has attracted considerable attention is sulforaphane (SFN), which exhibits anti-cancer, anti-diabetic, and anti-microbial properties. The present study was undertaken to assess effect of SFN alone and in combination with a chemotherapeutic agent, gemcitabine, on the proliferative potential of MCF-7 cells by cell viability assay and authenticated the results by nuclear morphological examination. Further we analyzed the modulation of expression of Bcl-2 and COX-2 on treatment of these cells with SFN by RT-PCR. SFN showed cytotoxic effects on MCF-7 cells in a dose- and time-dependent manner via an apoptotic mode of cell death. In addition, a combinational treatment of SFN and gemcitabine on MCF-7 cells resulted in growth inhibition in a synergistic manner with a combination index (CI) <1. Notably, SFN was found to significantly downregulate the expression of Bcl-2, an anti-apoptotic gene, and COX-2, a gene involved in inflammation, in a time-dependent manner. These results indicate that SFN induces apoptosis and anti-inflammatory effects on MCF-7 cells via downregulation of Bcl-2 and COX-2 respectively. The combination of SFN and gemcitabine may potentiate the efficacy of gemcitabine and minimize the toxicity to normal cells. Taken together, SFN may be a potent anti-cancer agent for breast cancer treatment.
Collapse
Affiliation(s)
- Arif Hussain
- Department of Biotechnology, Manipal University, Dubai, United Arab Emirates E-mail :
| | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Liang S, Peng X, Li X, Yang P, Xie L, Li Y, Du C, Zhang G. Silencing of CXCR4 sensitizes triple-negative breast cancer cells to cisplatin. Oncotarget 2015; 6:1020-30. [PMID: 25544759 PMCID: PMC4359214 DOI: 10.18632/oncotarget.2741] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 11/08/2014] [Indexed: 02/05/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer for which there is no effective treatment. Previously, we and others demonstrated that CXCR4 surface expression is an independent prognostic factor for disease relapse and survival in breast cancer. In this study, we investigated the effects of CXCR4 gene silencing on cisplatin chemosensitivity in human triple-negative breast cancer cell lines. We found that CXCR4 silencing significantly inhibited cell growth, decreased colony formation, and enhanced cisplatin sensitivity while overexpression of CXCR4 rendered cells more resistant to cisplatin. Moreover, the percentage of apoptosis and cell cycle arrest at the G2/M phase of cisplatin-treated CXCR4 knockdown cells was significantly higher than control cells. Furthermore, we demonstrated CXCR4 knockdown cells showed lower levels of mutant p53 and Bcl-2 protein than the control group, while also having higher levels of caspase-3 and Bax. However overexpression of CXCR4 had the reverse effect. In vivo experiments confirmed that downregulation of CXCR4 enhanced cisplatin anticancer activity in tumor-bearing mice, and that this enhanced anticancer activity is attributable to tumor cell apoptosis. Thus, this study indicates that CXCR4 can modulate cisplatin sensitivity in TNBC cells and suggests that CXCR4 may be a therapeutic target for TNBC.
Collapse
Affiliation(s)
- Sixian Liang
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| | - Xun Peng
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| | - Xiaoli Li
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| | - Ping Yang
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| | - Linhao Xie
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| | - Yaochen Li
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| | - Caiwen Du
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| | - Guojun Zhang
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515031, PR China
| |
Collapse
|
72
|
Tomao F, Papa A, Zaccarelli E, Rossi L, Caruso D, Minozzi M, Vici P, Frati L, Tomao S. Triple-negative breast cancer: new perspectives for targeted therapies. Onco Targets Ther 2015; 8:177-93. [PMID: 25653541 PMCID: PMC4303459 DOI: 10.2147/ott.s67673] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is a heterogeneous disease, encompassing a large number of entities showing different morphological features and having clinical behaviors. It has became apparent that this diversity may be justified by distinct patterns of genetic, epigenetic, and transcriptomic aberrations. The identification of gene-expression microarray-based characteristics has led to the identification of at least five breast cancer subgroups: luminal A, luminal B, normal breast-like, human epidermal growth factor receptor 2, and basal-like. Triple-negative breast cancer is a complex disease diagnosed by immunohistochemistry, and it is characterized by malignant cells not expressing estrogen receptors or progesterone receptors at all, and human epidermal growth factor receptor 2. Along with this knowledge, recent data show that triple-negative breast cancer has specific molecular features that could be possible targets for new biological targeted drugs. The aim of this article is to explore the use of new drugs in this particular setting, which is still associated with poor prognosis and high risk of distant recurrence and death.
Collapse
Affiliation(s)
- Federica Tomao
- Department of Gynecology and Obstetrics, “Sapienza” University of Rome, Policlinico “Umberto I”, Rome, Italy
| | - Anselmo Papa
- Department of Medico-Surgical Sciences and Biotechnologies, “Sapienza” University of Rome, Oncology Unit, Istituto Chirurgico Ortopedico Traumatologico, Latina, Italy
| | - Eleonora Zaccarelli
- Department of Medico-Surgical Sciences and Biotechnologies, “Sapienza” University of Rome, Oncology Unit, Istituto Chirurgico Ortopedico Traumatologico, Latina, Italy
| | - Luigi Rossi
- Department of Medico-Surgical Sciences and Biotechnologies, “Sapienza” University of Rome, Oncology Unit, Istituto Chirurgico Ortopedico Traumatologico, Latina, Italy
| | - Davide Caruso
- Department of Medico-Surgical Sciences and Biotechnologies, “Sapienza” University of Rome, Oncology Unit, Istituto Chirurgico Ortopedico Traumatologico, Latina, Italy
| | - Marina Minozzi
- Department of Medico-Surgical Sciences and Biotechnologies, “Sapienza” University of Rome, Oncology Unit, Istituto Chirurgico Ortopedico Traumatologico, Latina, Italy
| | - Patrizia Vici
- Division of Medical Oncology B, Regina Elena National Cancer Institute, Rome, Italy
| | - Luigi Frati
- Department of Molecular Medicine, “Sapienza” University of Rome, Policlinico “Umberto I”, Rome, Italy
| | - Silverio Tomao
- Department of Medico-Surgical Sciences and Biotechnologies, “Sapienza” University of Rome, Oncology Unit, Istituto Chirurgico Ortopedico Traumatologico, Latina, Italy
| |
Collapse
|
73
|
Capdevila-Busquets E, Badiola N, Arroyo R, Alcalde V, Soler-López M, Aloy P. Breast cancer genes PSMC3IP and EPSTI1 play a role in apoptosis regulation. PLoS One 2015; 10:e0115352. [PMID: 25590583 PMCID: PMC4295872 DOI: 10.1371/journal.pone.0115352] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 11/22/2014] [Indexed: 11/19/2022] Open
Abstract
A key element to delineate the biology of individual tumors is the regulation of apoptosis. In this work, we functionally characterize two breast cancer associated genes, the proteasome 26S subunit ATPase 3 interacting protein (PSMC3IP) and the epithelial-stromal interaction 1 (EPSTI1), to explore their potential apoptotic role in breast cancer. We first explore the existence of direct physical interactions with annotated BC-apoptotic genes. Based on the generated interaction network, we examine several apoptotic markers to determine the effect of PSMC3IP and EPSTI1 gene expression modulation in two different human breast cancer cell lines to suggest potential molecular mechanisms to unveil their role in the disease. Our results show that PSMC3IP and EPSTI1 are able to modulate the extrinsic apoptotic pathway in estrogen receptor positive and triple negative breast cancer cell lines, highlighting them as potential therapeutic targets.
Collapse
Affiliation(s)
- Eva Capdevila-Busquets
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Catalonia, Spain
| | - Nahuai Badiola
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Catalonia, Spain
| | - Rodrigo Arroyo
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Catalonia, Spain
| | - Víctor Alcalde
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Catalonia, Spain
| | - Montserrat Soler-López
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Catalonia, Spain
| | - Patrick Aloy
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
- * E-mail:
| |
Collapse
|
74
|
Kim JS, Park IH, Lee KS, Ro J. Outcomes of palliative weekly low-dose gemcitabine-Cisplatin chemotherapy in anthracycline- and taxane- pretreated metastatic breast cancer patients. J Breast Cancer 2014; 17:339-43. [PMID: 25548581 PMCID: PMC4278052 DOI: 10.4048/jbc.2014.17.4.339] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 12/03/2014] [Indexed: 11/30/2022] Open
Abstract
Purpose The combination of gemcitabine and cisplatin (GP) has been shown to be safe and efficacious for patients with metastatic breast cancer (MBC), pretreated with anthracyclines and taxanes. We assessed the efficacy and safety of weekly low-dose GP in patients with MBC. Methods We collected clinicopathological data from MBC patients who had been treated with gemcitabine, 800 mg/m2 plus cisplatin, 30 mg/m2 intravenously, on days 1 and 8 every 3 weeks, between January 2001 and November 2011 in Korea. Results The analysis included 294 patients previously treated anthracycline-xand taxane-based chemotherapies prior to GP (median age, 48 years [range, 28-78 years]; median follow-up duration, 63.9 months). Seventeen patients (5.8%) discontinued GP because of toxicities. The median progression-free survival (PFS) was 3.9 months (95% confidence interval [CI], 3.394.4 months) and the median overall survival (OS) was 27.7 months (95% CI, 17.6-37.8 months) months. Statistically significant factors for PFS were performance status (Eastern Cooperative Oncology Group, ≥2 vs. <2; hazard ratio [HR], 1.37; 95% CI, 1.02-1.85; p=0.037), distant disease-free interval (DDFI; ≤2 years vs. >2 years; HR, 1.66; 95% CI, 1.28-1.95, p<0.001), time interval from the diagnosis of metastasis to GP therapy (≤1 year vs. >1 year; HR, 1.48; 95% CI, 1.13-1.95, p<0.001), and presence of brain metastasis (HR, 1.47; 95% CI, 1.03-2.10; p=0.031). Similarly, DDFI (≤2 years vs. >2 years; HR, 2.07; 95% CI, 1.36-3.14; p<0.001) and the presence of brain metastasis (HR, 2.14; 95% CI, 1.27-3.61; p=0.004) were important factors for OS after GP treatment. Conclusion Weekly low-dose GP chemotherapy appears safe and effective for heavily pretreated MBC patients.
Collapse
Affiliation(s)
- Jung Sun Kim
- Center for Breast Cancer, National Cancer Center, Goyang, Korea
| | - In Hae Park
- Center for Breast Cancer, National Cancer Center, Goyang, Korea
| | - Keun Seok Lee
- Center for Breast Cancer, National Cancer Center, Goyang, Korea
| | - Jungsil Ro
- Center for Breast Cancer, National Cancer Center, Goyang, Korea
| |
Collapse
|
75
|
Karpel-Massler G, Pareja F, Aimé P, Shu C, Chau L, Westhoff MA, Halatsch ME, Crary JF, Canoll P, Siegelin MD. PARP inhibition restores extrinsic apoptotic sensitivity in glioblastoma. PLoS One 2014; 9:e114583. [PMID: 25531448 PMCID: PMC4273972 DOI: 10.1371/journal.pone.0114583] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 11/11/2014] [Indexed: 01/23/2023] Open
Abstract
Background Resistance to apoptosis is a paramount issue in the treatment of Glioblastoma (GBM). We show that targeting PARP by the small molecule inhibitors, Olaparib (AZD-2281) or PJ34, reduces proliferation and lowers the apoptotic threshold of GBM cells in vitro and in vivo. Methods The sensitizing effects of PARP inhibition on TRAIL-mediated apoptosis and potential toxicity were analyzed using viability assays and flow cytometry in established GBM cell lines, low-passage neurospheres and astrocytes in vitro. Molecular analyses included western blots and gene silencing. In vivo, effects on tumor growth were examined in a murine subcutaneous xenograft model. Results The combination treatment of PARP inhibitors and TRAIL led to an increased cell death with activation of caspases and inhibition of formation of neurospheres when compared to single-agent treatment. Mechanistically, pharmacological PARP inhibition elicited a nuclear stress response with up-regulation of down-stream DNA-stress response proteins, e.g., CCAAT enhancer binding protein (C/EBP) homology protein (CHOP). Furthermore, Olaparib and PJ34 increased protein levels of DR5 in a concentration and time-dependent manner. In turn, siRNA-mediated suppression of DR5 mitigated the effects of TRAIL/PARP inhibitor-mediated apoptosis. In addition, suppression of PARP-1 levels enhanced TRAIL-mediated apoptosis in malignant glioma cells. Treatment of human astrocytes with the combination of TRAIL/PARP inhibitors did not cause toxicity. Finally, the combination treatment of TRAIL and PJ34 significantly reduced tumor growth in vivo when compared to treatment with each agent alone. Conclusions PARP inhibition represents a promising avenue to overcome apoptotic resistance in GBM.
Collapse
Affiliation(s)
- Georg Karpel-Massler
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Fresia Pareja
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Pascaline Aimé
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Chang Shu
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Lily Chau
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | | | - John F Crary
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Peter Canoll
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Markus D Siegelin
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| |
Collapse
|
76
|
O'Shaughnessy J, Schwartzberg L, Danso MA, Miller KD, Rugo HS, Neubauer M, Robert N, Hellerstedt B, Saleh M, Richards P, Specht JM, Yardley DA, Carlson RW, Finn RS, Charpentier E, Garcia-Ribas I, Winer EP. Phase III study of iniparib plus gemcitabine and carboplatin versus gemcitabine and carboplatin in patients with metastatic triple-negative breast cancer. J Clin Oncol 2014; 32:3840-7. [PMID: 25349301 DOI: 10.1200/jco.2014.55.2984] [Citation(s) in RCA: 226] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
PURPOSE There is a lack of treatments providing survival benefit for patients with metastatic triple-negative breast cancer (mTNBC), with no standard of care. A randomized phase II trial showed significant benefit for gemcitabine, carboplatin, and iniparib (GCI) over gemcitabine and carboplatin (GC) in clinical benefit rate, response rate, progression-free survival (PFS), and overall survival (OS). Here, we formally compare the efficacy of these regimens in a phase III trial. PATIENTS AND METHODS Patients with stage IV/locally recurrent TNBC who had received no more than two previous chemotherapy regimens for mTNBC were randomly allocated to gemcitabine 1,000 mg/m(2) and carboplatin area under the curve 2 (days 1 and 8) alone or GC plus iniparib 5.6 mg/kg (days 1, 4, 8, and 11) every 3 weeks. Random assignment was stratified by the number of prior chemotherapies. The coprimary end points were OS and PFS. Patients receiving GC could cross over to iniparib on progression. RESULTS Five hundred nineteen patients were randomly assigned (261 GCI; 258 GC). In the primary analysis, no statistically significant difference was observed for OS (hazard ratio [HR] = 0.88; 95% CI, 0.69 to 1.12; P = .28) nor PFS (HR = 0.79; 95% CI, 0.65 to 0.98; P = .027). An exploratory analysis showed that patients in the second-/third-line had improved OS (HR = 0.65; 95% CI, 0.46 to 0.91) and PFS (HR = 0.68; 95% CI, 0.50 to 0.92) with GCI. The safety profile for GCI was similar to GC. CONCLUSION The trial did not meet the prespecified criteria for the coprimary end points of PFS and OS in the ITT population. The potential benefit with iniparib observed in second-/third-line subgroup warrants further evaluation.
Collapse
Affiliation(s)
- Joyce O'Shaughnessy
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA.
| | - Lee Schwartzberg
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Michael A Danso
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Kathy D Miller
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Hope S Rugo
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Marcus Neubauer
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Nicholas Robert
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Beth Hellerstedt
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Mansoor Saleh
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Paul Richards
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Jennifer M Specht
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Denise A Yardley
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Robert W Carlson
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Richard S Finn
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Eric Charpentier
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Ignacio Garcia-Ribas
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Eric P Winer
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
77
|
Sonnenblick A, de Azambuja E, Azim HA, Piccart M. An update on PARP inhibitors--moving to the adjuvant setting. Nat Rev Clin Oncol 2014; 12:27-41. [PMID: 25286972 DOI: 10.1038/nrclinonc.2014.163] [Citation(s) in RCA: 274] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Inhibition of poly(ADP-ribose) polymerase (PARP) enzymes is a potential synthetic lethal therapeutic strategy in cancers harbouring specific DNA-repair defects, including those arising in carriers of BRCA1 or BRCA2 mutations. Since the development of first-generation PARP inhibitors more than a decade ago, numerous clinical trials have been performed to validate their safety and efficacy, bringing us to the stage at which adjuvant therapy with PARP inhibitors is now being considered as a viable treatment option for patients with breast cancer. Nevertheless, the available data do not provide clear proof that these drugs are efficacious in the setting of metastatic disease. Advancement of a therapy to the neoadjuvant and adjuvant settings without such evidence is exceptional, but seems reasonable in the case of PARP inhibitors because the target population that might benefit from this class of drugs is small and well defined. This Review describes the evolution of PARP inhibitors from bench to bedside, and provides an up-to-date description of the key published or otherwise reported clinical trials of these agents. The specific considerations and challenges that might be encountered when implementing these compounds in the adjuvant treatment of breast cancer in the clinic are also highlighted.
Collapse
Affiliation(s)
- Amir Sonnenblick
- BrEAST Data Centre, Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo 125, B-1000 Brussels, Belgium
| | - Evandro de Azambuja
- BrEAST Data Centre, Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo 125, B-1000 Brussels, Belgium
| | - Hatem A Azim
- BrEAST Data Centre, Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo 125, B-1000 Brussels, Belgium
| | - Martine Piccart
- BrEAST Data Centre, Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo 125, B-1000 Brussels, Belgium
| |
Collapse
|
78
|
Clinical Evaluation of Platinum Agents for the Treatment of Triple Negative Breast Cancer. CURRENT BREAST CANCER REPORTS 2014. [DOI: 10.1007/s12609-014-0160-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
79
|
Alli E, Solow-Cordero D, Casey SC, Ford JM. Therapeutic targeting of BRCA1-mutated breast cancers with agents that activate DNA repair. Cancer Res 2014; 74:6205-15. [PMID: 25217519 DOI: 10.1158/0008-5472.can-14-1716] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancers due to germline mutations in the BRCA1 gene tend to lack targets for approved chemoprevention agents. This study aimed at a targeted chemoprevention strategy for BRCA1-associated malignancies. Mutant BRCA1 limits the base-excision DNA repair activity that addresses oxidative DNA damage, the accumulation of which heightens one's risk for cancer. Therefore, we conducted a high-throughput chemical screen to identify drug candidates that could attenuate the inhibitory effects of mutant BRCA1 on this repair activity, thereby describing a new class of DNA repair-activating chemopreventive agents. In the screen design, such drugs functioned by enhancing base-excision DNA repair of oxidative DNA damage in the presence of mutant BRCA1, with minimal cytotoxicity. We identified at least one new agent that decreased malignant properties associated with tumorigenesis, including anchorage-independent growth and tumor progression. This work offers a preclinical proof-of-concept for a wholly new approach to chemoprevention in carriers of BRCA1 mutations as a strategy to reduce the prevalence of BRCA1-associated malignancy.
Collapse
Affiliation(s)
- Elizabeth Alli
- Department of Medicine/Oncology, Stanford University School of Medicine, Stanford, California
| | - David Solow-Cordero
- Department of Chemical and Systems Biology and Stanford High-Throughput Bioscience Center, Stanford University School of Medicine, Stanford, California
| | - Stephanie C Casey
- Department of Medicine/Oncology, Stanford University School of Medicine, Stanford, California
| | - James M Ford
- Department of Medicine/Oncology, Stanford University School of Medicine, Stanford, California. Department of Genetics, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
80
|
Bryant C, Rawlinson R, Massey AJ. Chk1 inhibition as a novel therapeutic strategy for treating triple-negative breast and ovarian cancers. BMC Cancer 2014; 14:570. [PMID: 25104095 PMCID: PMC4137066 DOI: 10.1186/1471-2407-14-570] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 07/28/2014] [Indexed: 12/31/2022] Open
Abstract
Background Chk1 inhibitors are currently in clinical trials as putative potentiators of cytotoxic chemotherapy drugs. Chk1 inhibitors may exhibit single agent anti-tumor activity in cancers with underlying DNA repair, DNA damage response or DNA replication defects. Methods Here we describe the cellular effects of the pharmacological inhibition of the checkpoint kinase Chk1 by the novel inhibitor V158411 in triple-negative breast cancer and ovarian cancer. Cytotoxicity, the effect on DNA damage response and cell cycle along with the ability to potentiate gemcitabine and cisplatin cytotoxicity in cultured cells was investigated. Western blotting of proteins involved in DNA repair, checkpoint activation, cell cycle and apoptosis was used to identify potential predictive biomarkers of Chk1 inhibitor sensitivity. Results The Chk1 inhibitors V158411, PF-477736 and AZD7762 potently inhibited the proliferation of triple-negative breast cancer cells as well as ovarian cancer cells, and these cell lines were sensitive compared to ER positive breast and other solid cancer cells lines. Inhibition of Chk1 in these sensitive cell lines induced DNA damage and caspase-3/7 dependent apoptosis. Western blot profiling identified pChk1 (S296) as a predictive biomarker of Chk1 inhibitor sensitivity in ovarian and triple-negative breast cancer and pH2AX (S139) in luminal breast cancer. Conclusions This finding suggests that Chk1 inhibitors either as single agents or in combination chemotherapy represents a viable therapeutic option for the treatment of triple-negative breast cancer. pChk1 (S296) tumor expression levels could serve as a useful biomarker to stratify patients who might benefit from Chk1 inhibitor therapy.
Collapse
|
81
|
Liu L, Zhou W, Cheng CT, Ren X, Somlo G, Fong MY, Chin AR, Li H, Yu Y, Xu Y, O'Connor STF, O'Connor TR, Ann DK, Stark JM, Wang SE. TGFβ induces "BRCAness" and sensitivity to PARP inhibition in breast cancer by regulating DNA-repair genes. Mol Cancer Res 2014; 12:1597-609. [PMID: 25103497 DOI: 10.1158/1541-7786.mcr-14-0201] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
UNLABELLED Transforming growth factor beta (TGFβ) proteins are multitasking cytokines, in which high levels at tumor sites generally correlate with poor prognosis in human patients with cancer. Previously, it was reported that TGFβ downregulates the expression of ataxia telangiectasia-mutated (ATM) and mutS homolog 2 (MSH2) in breast cancer cells through an miRNA-mediated mechanism. In this study, expression of a panel of DNA-repair genes was examined, identifying breast cancer 1, early onset (BRCA1) as a target downregulated by TGFβ through the miR181 family. Correlations between the expression levels of TGFβ1 and the miR181/BRCA1 axis were observed in primary breast tumor specimens. By downregulating BRCA1, ATM, and MSH2, TGFβ orchestrates DNA damage response in certain breast cancer cells to induce a "BRCAness" phenotype, including impaired DNA-repair efficiency and synthetic lethality to the inhibition of poly (ADP-ribose) polymerase (PARP). Xenograft tumors with active TGFβ signaling exhibited resistance to the DNA-damaging agent doxorubicin but increased sensitivity to the PARP inhibitor ABT-888. Combination of doxorubicin with ABT-888 significantly improved the treatment efficacy in TGFβ-active tumors. Thus, TGFβ can induce "BRCAness" in certain breast cancers carrying wild-type BRCA genes and enhance the responsiveness to PARP inhibition, and the molecular mechanism behind this is characterized. IMPLICATIONS These findings enable better selection of patients with sporadic breast cancer for PARP interventions, which have exhibited beneficial effects in patients carrying BRCA mutations.
Collapse
Affiliation(s)
- Liang Liu
- Department of Cancer Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Biotherapy and Key Laboratory of Cancer Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Weiying Zhou
- Department of Cancer Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Pharmacology, College of Pharmacy, The Third Military Medical University, Chongqing, China
| | - Chun-Ting Cheng
- Department of Molecular Pharmacology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. City of Hope Irell and Manella Graduate School of Biological Sciences, Duarte, California
| | - Xiubao Ren
- Department of Biotherapy and Key Laboratory of Cancer Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - George Somlo
- Department of Medical Oncology, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Miranda Y Fong
- Department of Cancer Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Andrew R Chin
- Department of Cancer Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. City of Hope Irell and Manella Graduate School of Biological Sciences, Duarte, California
| | - Hui Li
- Department of Biotherapy and Key Laboratory of Cancer Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yang Yu
- Department of Biotherapy and Key Laboratory of Cancer Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yang Xu
- Department of Cancer Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | | | - Timothy R O'Connor
- Department of Cancer Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - David K Ann
- Department of Molecular Pharmacology, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Jeremy M Stark
- Department of Radiation Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Shizhen Emily Wang
- Department of Cancer Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Biotherapy and Key Laboratory of Cancer Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| |
Collapse
|
82
|
Dual inhibition of REV-ERBβ and autophagy as a novel pharmacological approach to induce cytotoxicity in cancer cells. Oncogene 2014; 34:2597-608. [PMID: 25023698 PMCID: PMC4761647 DOI: 10.1038/onc.2014.203] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 05/16/2014] [Accepted: 06/06/2014] [Indexed: 12/24/2022]
Abstract
REV-ERBα and REV-ERBβ nuclear receptors regulate several physiological processes, including circadian rhythm and metabolism. A previous study reported the REV-ERBα gene to be co-overexpressed with ERBB2 in breast cancer cell lines. Surprisingly, we found that several tumor types, including a number of breast cancer cell lines, predominantly express the REV-ERBβ variant. This pattern was independent of ERBB2 and ER status, and opposite to that of non-cancer mammary epithelial HMEC cells, in which REV-ERBα was the major variant. Consistent with this molecular profile, REV-ERB target genes in both circadian and metabolic pathways were derepressed upon silencing of REV-ERBβ, but not REV-ERBα. Strikingly, we found that REV-ERBβ is a determinant of sensitivity to chloroquine, a clinically relevant lysosomotropic agent that suppresses autophagy. The cytoprotective function of REV-ERBβ appears to operate downstream of autophagy blockade. Through compound screening, we identified ARN5187, a novel lysosomotropic REV-ERBβ ligand with a dual inhibitory activity toward REV-ERB-mediated transcriptional regulation and autophagy. Remarkably, although ARN5187 and chloroquine share similar lysosomotropic potency and have a similar effect on autophagy inhibition, ARN5187 is significantly more cytotoxic. Collectively, our results reveal that dual inhibition of REV-ERBβ and autophagy is an effective strategy for eliciting cytotoxicity in cancer cells. Furthermore, our discovery of a novel inhibitor compound of both REV-ERB and autophagy may provide a scaffold for the discovery of new multifunctional anticancer agents.
Collapse
|
83
|
Afghahi A, Telli ML. The role of platinum therapy in triple-negative breast cancer. BREAST CANCER MANAGEMENT 2014. [DOI: 10.2217/bmt.14.21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY Breast cancer is a heterogeneous disease consisting of distinct biological subtypes with therapeutic and prognostic implications. Triple-negative breast cancer (TNBC) often follows a more aggressive disease course with poorer disease-specific survival compared with other breast cancer subtypes. Despite tremendous efforts to change the current treatment algorithm for women with TNBC, little has changed in over a decade. Encouraging results emerging from elegant preclinical studies to early-phase clinical trials demonstrate that platinum agents may have a role to play in the treatment of TNBC. In addition to germline BRCA1 and BRCA2 mutation status, other biomarkers with the ability to assess platinum responsiveness are emerging, including tissue-based assays that detect genomic 'scarring' caused by accumulated DNA damage and immunological biomarkers. Prospective evaluation of these biomarkers in a clinical setting is a high priority, as tailoring therapy with the incorporation of platinum agents based on biomarkers of response is an intriguing alternative to current standard of care.
Collapse
Affiliation(s)
- Anosheh Afghahi
- Stanford University School of Medicine, Division of Medical Oncology, Stanford, CA, USA
| | - Melinda L Telli
- Stanford University School of Medicine, Division of Medical Oncology, Stanford, CA, USA
| |
Collapse
|
84
|
Murai J, Zhang Y, Morris J, Ji J, Takeda S, Doroshow JH, Pommier Y. Rationale for poly(ADP-ribose) polymerase (PARP) inhibitors in combination therapy with camptothecins or temozolomide based on PARP trapping versus catalytic inhibition. J Pharmacol Exp Ther 2014; 349:408-16. [PMID: 24650937 PMCID: PMC4019318 DOI: 10.1124/jpet.113.210146] [Citation(s) in RCA: 228] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 03/10/2014] [Indexed: 01/20/2023] Open
Abstract
We recently showed that poly(ADP-ribose) polymerase (PARP) inhibitors exert their cytotoxicity primarily by trapping PARP-DNA complexes in addition to their NAD(+)-competitive catalytic inhibitory mechanism. PARP trapping is drug-specific, with olaparib exhibiting a greater ability than veliparib, whereas both compounds are potent catalytic PARP inhibitors. Here, we evaluated the combination of olaparib or veliparib with therapeutically relevant DNA-targeted drugs, including the topoisomerase I inhibitor camptothecin, the alkylating agent temozolomide, the cross-linking agent cisplatin, and the topoisomerase II inhibitor etoposide at the cellular and molecular levels. We determined PARP-DNA trapping and catalytic PARP inhibition in genetically modified chicken lymphoma DT40, human prostate DU145, and glioblastoma SF295 cancer cells. For camptothecin, both PARP inhibitors showed highly synergistic effects due to catalytic PARP inhibition, indicating the value of combining either veliparib or olaparib with topoisomerase I inhibitors. On the other hand, for temozolomide, PARP trapping was critical in addition to catalytic inhibition, consistent with the fact that olaparib was more effective than veliparib in combination with temozolomide. For cisplatin and etoposide, olaparib only showed no or a weak combination effect, which is consistent with the lack of involvement of PARP in the repair of cisplatin- and etoposide-induced lesions. Hence, we conclude that catalytic PARP inhibitors are highly effective in combination with camptothecins, whereas PARP inhibitors capable of PARP trapping are more effective with temozolomide. Our study provides insights in combination treatment rationales for different PARP inhibitors.
Collapse
Affiliation(s)
- Junko Murai
- Developmental Therapeutics Branch, Laboratory of Molecular Pharmacology, Center for Cancer Research (Ju.M., J.H.D., Y.P.), National Clinical Target Validation Laboratory (Y.Z., J.J.), and Division of Cancer Treatment and Diagnosis (Jo.M., J.H.D.), National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Yoshidakonoe, Sakyo-ku, Kyoto, Japan (Ju.M., S.T.)
| | | | | | | | | | | | | |
Collapse
|
85
|
Jørgensen CLT, Nielsen TO, Bjerre KD, Liu S, Wallden B, Balslev E, Nielsen DL, Ejlertsen B. PAM50 breast cancer intrinsic subtypes and effect of gemcitabine in advanced breast cancer patients. Acta Oncol 2014; 53:776-87. [PMID: 24359601 DOI: 10.3109/0284186x.2013.865076] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND In vitro studies suggest basal breast cancers are more sensitive to gemcitabine relative to other intrinsic subtypes. The main objective of this study was to use specimens from a randomized clinical trial to evaluate whether the basal-like subtype identifies patients with advanced breast cancer who benefit from gemcitabine plus docetaxel (GD) compared to single agent docetaxel (D). MATERIAL AND METHODS From patients randomly assigned to GD or D, RNA was isolated from archival formalin-fixed, paraffin-embedded primary breast tumor tissue and used for PAM50 intrinsic subtyping by NanoString nCounter. Statistical analyses were prespecified as a formal prospective-retrospective clinical trial correlative study. Using time to progression (TTP) as primary endpoint, overall survival (OS) and response rate as secondary endpoints, relationships between subtypes and outcome after chemotherapy were analyzed by the Kaplan-Meier method, and Cox proportional hazards regression models. Data analysis was performed independently by the Danish Breast Cancer Cooperative Group (DBCG) statistical core and all statistical tests were two-sided. RESULTS RNA from 270 patients was evaluable; 84 patients (31%) were classified as luminal A, 97 (36%) luminal B, 43 (16%) basal-like, and 46 (17%) as HER2-enriched. PAM50 intrinsic subtype was a significant independent prognostic factor for both TTP (p=0.014) and OS (p=0.0003). Response rate was not different by subtype, and PAM50 was not a predictor of TTP by treatment arm. PAM50 was however a highly significant predictor of OS following GD compared to D (pinteraction=0.0016). Patients with a basal-like subtype had a significant reduction in OS events [hazard ratio (HR)=0.29, 95% confidence interval (CI)=0.15-0.57; pinteraction=0.0006]. CONCLUSION A significantly improved and clinically important prolongation of survival was seen from the addition of gemcitabine to docetaxel in advanced basal-like breast cancer patients.
Collapse
|
86
|
Yadav BS, Sharma SC, Chanana P, Jhamb S. Systemic treatment strategies for triple-negative breast cancer. World J Clin Oncol 2014; 5:125-133. [PMID: 24829859 PMCID: PMC4014784 DOI: 10.5306/wjco.v5.i2.125] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Revised: 12/25/2013] [Accepted: 01/17/2014] [Indexed: 02/06/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is defined by the lack of immunohistochemical expression of the estrogen and progesterone receptors and human epidermal growth factor receptor 2 (EGFR2). Most TNBC has a basal-like molecular phenotype by gene expression profiling and shares clinical and pathological features with hereditary BRCA1 related breast cancers. This review evaluates the activity of available chemotherapy and targeted agents in TNBC. A systematic review of PubMed and conference databases was carried out to identify randomised clinical trials reporting outcomes in women with TNBC treated with chemotherapy and targeted agents. Our review identified TNBC studies of chemotherapy and targeted agents with different mechanisms of action, including induction of synthetic lethality and inhibition of angiogenesis, growth and survival pathways. TNBC is sensitive to taxanes and anthracyclins. Platinum agents are effective in TNBC patients with BRCA1 mutation, either alone or in combination with poly adenosine diphosphate polymerase 1 inhibitors. Combinations of ixabepilone and capecitabine have added to progression-free survival (PFS) without survival benefit in metastatic TNBC. Antiangiogenic agents, tyrosine kinase inhibitors and EGFR inhibitors in combination with chemotherapy produced only modest gains in PFS and had little impact on survival. TNBC subgroups respond differentially to specific targeted agents. In future, the treatment needs to be tailored for a specific patient, depending on the molecular characteristics of their malignancy. TNBC being a chemosensitive entity, combination with targeted agents have not produced substantial improvements in outcomes. Appropriate patient selection with rationale combinations of targeted agents is needed for success.
Collapse
|
87
|
|
88
|
Pierce A, McGowan PM, Cotter M, Mullooly M, O'Donovan N, Rani S, O'Driscoll L, Crown J, Duffy MJ. Comparative antiproliferative effects of iniparib and olaparib on a panel of triple-negative and non-triple-negative breast cancer cell lines. Cancer Biol Ther 2014; 14:537-45. [PMID: 23760496 DOI: 10.4161/cbt.24349] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
PARP inhibitors, both as monotherapy and in combination with cytotoxic drugs, are currently undergoing clinical trials in several different cancer types. In this investigation, we compared the antiproliferative activity of two PARP/putative PARP inhibitors, i.e., olaparib and iniparib, in a panel of 14 breast cancer cell lines (seven tripe-negative and seven non-triple-negative). In almost all cell lines investigated, olaparib was a more potent inhibitor of cell growth than iniparib. Inhibition by both drugs was cell line-dependent and independent of the molecular subtype status of the cells, i.e., whether cells were triple-negative or non-triple negative. Although the primary target of PARP inhibitors is PARP1, no significant association was found between baseline levels of PARP1 activity and inhibition with either agent. Similarly, no significant correlation was evident between sensitivity and levels of CDK1, BRCA1 or miR-182. Combined addition of olaparib and either the CDK1 inhibitor, RO-3306 or a pan HER inhibitor (neratinib, afatinib) resulted in superior growth inhibition to that obtained with olaparib alone. We conclude that olaparib, in contrast to iniparib, is a strong inhibitor of breast cancer cell growth and may have efficacy in breast cancer irrespective of its molecular subtype, i.e., whether HER2-positive, estrogen receptor (ER)-positive or triple-negative. Olaparib, in combination with a selective CDK1 inhibitor or a pan HER inhibitor, is a potential new approach for treating breast cancer.
Collapse
Affiliation(s)
- Aisling Pierce
- Education and Research Centre, St. Vincent's University Hospital, Dublin, Ireland
| | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Abstract
Although characterization of triple-negative breast cancer (TNBC) using mRNA gene expression profiling has certainly provided important insights, the concept of targeting DNA repair defects with DNA damaging therapeutics such as platinum in TNBC has been advanced from studies focusing on both germline and somatic genetic alterations associated with this breast cancer subtype. A growing body of preclinical and clinical data suggests that platinum chemotherapy has a potential role to play in the treatment of both early-stage and advanced TNBC, though results are not yet definitive. Randomized clinical trials that incorporate biomarkers of response, including germline BRCA1 and BRCA2 mutation status as well as tumor-based measures of genomic "scarring" resulting from the accumulation of DNA damage in tumors with deficient repair capacity, will help to clarify the optimal use and activity of platinum in TNBC.
Collapse
Affiliation(s)
- Melinda Telli
- From the Stanford University School of Medicine, Division of Oncology, Stanford, CA
| |
Collapse
|
90
|
Shah GM, Robu M, Purohit NK, Rajawat J, Tentori L, Graziani G. PARP Inhibitors in Cancer Therapy: Magic Bullets but Moving Targets. Front Oncol 2013; 3:279. [PMID: 24294592 PMCID: PMC3827545 DOI: 10.3389/fonc.2013.00279] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 10/29/2013] [Indexed: 02/03/2023] Open
Affiliation(s)
- Girish M Shah
- Laboratory for Skin Cancer Research, CHU-Q (CHUL) Research Centre, Laval University , Quebec City, QC , Canada
| | | | | | | | | | | |
Collapse
|
91
|
McPherson LA, Shen Y, Ford JM. Poly (ADP-ribose) polymerase inhibitor LT-626: Sensitivity correlates with MRE11 mutations and synergizes with platinums and irinotecan in colorectal cancer cells. Cancer Lett 2013; 343:217-23. [PMID: 24215868 DOI: 10.1016/j.canlet.2013.10.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/30/2013] [Accepted: 10/10/2013] [Indexed: 02/07/2023]
Abstract
Some colorectal cancers (CRC) display microsatellite instability (MSI) leading to mutations in genes such as MRE11. The aim of this study was to determine whether MSI or MRE11 mutational status correlates with sensitivity to the PARP inhibitor LT-626 and whether LT-626 synergizes with DNA-damaging chemotherapeutic agents. CRC cells harboring biallelic MRE11 mutations were more sensitive to LT-626 and stable overexpression or knock-down of MRE11 in cell lines correlated with sensitivity. Synergism was evident between LT-626 and cisplatin, oxaliplatin and SN-38 suggesting that PARP inhibitors in combination with DNA damaging agents may be a successful strategy for treatment of CRC.
Collapse
Affiliation(s)
- Lisa A McPherson
- Division of Oncology, Stanford University School of Medicine, Stanford, CA, United States
| | - Yuqiao Shen
- BioMarin Pharmaceutical Inc., Novato, CA, United States
| | - James M Ford
- Division of Oncology, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
92
|
Zhang P, Zheng BB, Wang HY, Chen JH, Liu XY, Guo XL. DLJ14, a novel chemo-sensitization agent, enhances therapeutic effects of adriamycin against MCF-7/A cells both in vitro and in vivo. ACTA ACUST UNITED AC 2013; 66:398-407. [PMID: 24533823 DOI: 10.1111/jphp.12168] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 09/24/2013] [Indexed: 12/01/2022]
Abstract
OBJECTIVES We investigated the chemo-sensitization of a ligustrazine derivate, (E)-2-(2, 4-dimethoxystyryl)-3, 5, 6-trimethylpyrazine (DLJ14) on Adriamycin (Adr, Wanle, Shenzhen, China)-resistant human breast cancer (MCF-7/A) cells both in vivo and in vitro. METHODS The antitumour effects of DLJ14 and Adr was observed in MCF-7/A cells by 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay in vitro and was evaluated by MCF-7/A xenografts in nude mice. The intracellular Adr accumulation was assessed by mean fluorescence intensity of Adr. The messenger RNA level of glutathione (GSH) S-transferase (GST)π in MCF-7/A cells was determined by real-time reverse transcription PCR assay. The expression of GSTπ, c-jun NH2 -terminal kinase (JNK) and phosphor-JNK (p-JNK) was detected by Western blotting method. KEY FINDINGS The MTT results showed that DLJ14 exhibited a weak inhibition on proliferation of both MCF-7 and MCF-7/A cells, in contrast with the strong inhibition of verapamil. When DLJ14 is combined with Adr, the inhibitory effect on MCF-7/A cells and MCF-7/A xenografts was enhanced significantly through increasing intracellular accumulation of Adr by inhibition of GSH level and the activity of GSH peroxidase and GST. Moreover, DLJ14 could downregulate the expression of GSTπ and increase the expression of JNK and p-JNK in MCF-7/A cells or in xenografts. CONCLUSION DLJ14 is a promising chemo-sensitization candidate for the reversal of multidrug resistance in cancers.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | | | | | | | | | | |
Collapse
|
93
|
Mateo J, Ong M, Tan DSP, Gonzalez MA, de Bono JS. Appraising iniparib, the PARP inhibitor that never was--what must we learn? Nat Rev Clin Oncol 2013; 10:688-96. [PMID: 24129347 DOI: 10.1038/nrclinonc.2013.177] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Several drugs targeting poly(ADP-ribose) polymerase (PARP) enzymes are under development. Responses have been observed in patients with germline mutations in BRCA1 and BRCA2, with further data supporting antitumour activity of PARP inhibitors in sporadic ovarian cancer. Strategies to identify other predictive biomarkers remain under investigation. Iniparib was purported to be a PARP inhibitor that showed promising results in randomized phase II trials in patients with triple-negative breast cancer. Negative results from a phase III study in this disease setting, however, tempered enthusiasm for this agent. Recently, data from in vitro experiments suggest that iniparib is not only structurally distinct from other described PARP inhibitors, but is also a poor inhibitor of PARP activity. In this context, the negative iniparib phase III data might have erroneously promulgated the notion that PARP inhibition is not an effective therapeutic strategy. Here, we scrutinize the development of iniparib from preclinical studies to registration trials, and identify and discuss the pitfalls in the development of anticancer drugs to prevent future late-stage trial failures.
Collapse
Affiliation(s)
- Joaquin Mateo
- Drug Development Unit, Division of Cancer Therapeutics and Division of Clinical Studies, The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, Downs Road, Sutton, Surrey SM2 5PT, UK
| | | | | | | | | |
Collapse
|
94
|
Prabhakaran P, Hassiotou F, Blancafort P, Filgueira L. Cisplatin induces differentiation of breast cancer cells. Front Oncol 2013; 3:134. [PMID: 23761858 PMCID: PMC3669802 DOI: 10.3389/fonc.2013.00134] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 05/13/2013] [Indexed: 12/25/2022] Open
Abstract
Breast tumors are heterogeneous including cells with stem cell properties and more differentiated cells. This heterogeneity is reflected into the molecular breast cancer subtypes. Breast cancer stem cells are resistant to chemotherapy, thus recent efforts are focusing on identifying treatments that shift them toward a more differentiated phenotype, making them more susceptible to chemotherapy. We examined whether the drug cisplatin induces differentiation in breast cancer cell lines that represent different breast cancer subtypes. We used three cell lines representing triple-negative breast cancers, BT-549 and MDA-MB-231 (claudin-low), and MDA-MB-468 (basal-like), along with estrogen and progesterone receptor positive MCF-7 cells (luminal). Cisplatin was applied at 2.5, 5, 10, and 20 μM, and cell viability and proliferation were measured using MTS and BrdU assays, respectively. The effect of cisplatin on the cellular hierarchy was examined by flow cytometry, immunofluorescence and qRT-PCR. Cisplatin treatment of 10 and 20 μM reduced cell viability by 36–51% and proliferation capacity by 36–67%. Treatment with cisplatin resulted in 12–67% down-regulation of stem cell markers (CD49f, SSEA4) and 10–130% up-regulation of differentiation markers (CK18, SMA, β-tubulin). At the mRNA level, CD49f was down-regulated whilst β-tubulin was up-regulated in the claudin-low cell lines. SSEA4 protein expression decreased upon cisplatin treatment, but SSEA4 mRNA expression increased indicating a differential regulation of cisplatin at the post-transcriptional level. It is concluded that cisplatin reduces breast cancer cell survival and induces differentiation of stem/progenitor cell subpopulations within breast cancer cell lines. These effects indicate the potential of this drug to target specific chemotherapy-resistant cells within a tumor.
Collapse
Affiliation(s)
- Praseetha Prabhakaran
- School of Anatomy, Physiology and Human Biology, The University of Western Australia , Crawley, Perth, WA , Australia ; Faculty of Biosciences and Bioengineering, Universiti Teknologi Malaysia , Skudai, Johor , Malaysia
| | | | | | | |
Collapse
|
95
|
Crown J, O'Shaughnessy J, Gullo G. Emerging targeted therapies in triple-negative breast cancer. Ann Oncol 2013; 23 Suppl 6:vi56-65. [PMID: 23012305 DOI: 10.1093/annonc/mds196] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Standard chemotherapy regimens can prove effective for patients with early triple-negative breast cancer (TNBC); however, patients with advanced disease typically respond poorly and rapidly progress, and the outcome is poor. New targeted therapies are therefore an urgent unmet medical need for this patient population. Translational and clinical studies into new TNBC treatments have been facilitated by the increased understanding of the aberrant signal transduction pathways regulating growth and survival and the development of chemoresistance in TNBC. Some of the established targeted agents that have been approved in other indications may prove beneficial to patients with TNBC; however, in the absence of approved targeted agents for the treatment of TNBC, most new agents remain experimental. Increased understanding of molecular profiles of TNBC subtypes is likely to improve therapeutic strategies with targeted agents. Novel strategies have reached clinical evaluation in patients with TNBC, including targeting angiogenesis vascular endothelial growth factor and proliferation signalling (receptor tyrosine kinases and mammalian target of rapamycin). Aggressive TNBCs have been found to associate closely with BRCA1 mutation or dysregulation. The recent development of new investigational agents targeting DNA repair, either directly with poly(adenosine disphosphate-ribose) polymerase inhibitors or indirectly through DNA-binding or DNA-damage potentiation, is a major focus of current clinical studies. These and other targeted therapies represent a new approach to TNBC therapy.
Collapse
Affiliation(s)
- J Crown
- St Vincent's University Hospital, Dublin, Ireland.
| | | | | |
Collapse
|
96
|
Mapping genetic alterations causing chemoresistance in cancer: identifying the roads by tracking the drivers. Oncogene 2013; 32:5315-30. [PMID: 23474753 DOI: 10.1038/onc.2013.48] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 01/16/2013] [Accepted: 01/16/2013] [Indexed: 12/12/2022]
Abstract
Although new agents are implemented to cancer therapy, we lack fundamental understandings of the mechanisms of chemoresistance, the main obstacle to cure in cancer. Here we review clinical evidence linking molecular defects to drug resistance across different tumour forms and discuss contemporary experimental evidence exploring these mechanisms. Although evidence, in general, is sparse and fragmentary, merging knowledge links drug resistance, and also sensitivity, to defects in functional pathways having a key role in cell growth arrest or death and DNA repair. As these pathways may act in concert, there is a need to explore multiple mechanisms in parallel. Taking advantage of massive parallel sequencing and other novel high-throughput technologies and base research on biological hypotheses, we now have the possibility to characterize functional defects related to these key pathways and to design a new generation of studies identifying the mechanisms controlling resistance to different treatment regimens in different tumour forms.
Collapse
|
97
|
p73 G4C14-to-A4T14 polymorphisms are positively correlated with triple-negative breast cancer in southwestern China. Med Oncol 2013; 30:515. [PMID: 23443851 DOI: 10.1007/s12032-013-0515-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Accepted: 02/16/2013] [Indexed: 10/27/2022]
Abstract
p73 gene shares structural and functional similarities to p53 and plays an important role in modulating cell cycle arrest and apoptosis. A common non-coding polymorphism of p73 G4C14-to-A4T14 (rs2273953 and rs1801173) at exon 2 may affect gene expression, thus, it may lead to functional significance. The correlation of this polymorphism with clinicopathologic variables of patients with breast cancer has not been investigated. In this study, single-nucleotide polymorphisms (SNPs) of p73 G4C14-to-A4T14 were genotyped by Sequenom MassArray-iPLEX GOLD System in 170 patients with breast cancer. Data were analyzed via t test, chi-square test, and logistic regression analysis. There was no significant correlation between p73 G4C14-to-A4T14 polymorphisms and the patient characteristics, such as clinical TNM stage, menopausal status, axillary lymph node metastasis, pathological type, estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2). No significant relationship was observed between the p73 G4C14-to-A4T14 polymorphism and p73 protein expression in cancer tissues. The frequency of GC/GC genotype in patients with triple-negative breast cancer (TNBC) was 78.9 %, that of patients with others was 57.6 %, and the difference had statistical significance (χ ( 2 ) = 5.74, P = 0.02). p73 G4C14-to-A4T14 polymorphisms were negatively correlated with chemosensitivity for anthracycline-based chemotherapy in breast cancer (P > 0.05). p73 G4C14-to-A4T14 polymorphisms are positively correlated with TNBC, and p73 gene may play a critical role in a novel therapeutic strategy to TNBC. Additional larger studies are required to test these hypotheses.
Collapse
|
98
|
Montariello D, Troiano A, Malanga M, Calabrò V, Quesada P. p63 involvement in poly(ADP-ribose) polymerase 1 signaling of topoisomerase I-dependent DNA damage in carcinoma cells. Biochem Pharmacol 2013; 85:999-1006. [PMID: 23376119 DOI: 10.1016/j.bcp.2013.01.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 01/21/2013] [Accepted: 01/22/2013] [Indexed: 10/27/2022]
Abstract
Poly(ADP-ribose)polymerase 1 (PARP-1) inhibitors are thought as breakthrough for cancer treatment in solid tumors such as breast cancer through their effects on PARP's enzymatic activity. Our previous findings showed that the hydrophilic PARP inhibitor PJ34 enhances the sensitivity of p53 proficient MCF7 breast carcinoma cells to topotecan, a DNA Topoisomerase I (TOP 1) inhibitor. In the present study, we combine the classical TOP 1 poison camptothecin or its water-soluble derivative topotecan with PJ34 to investigate the potentiation of chemotherapeutic efficiency in MCF7 (p53(WT)), MDA-MB231 (p53(mut)) breast carcinoma cells and SCC022 (p53(null)) squamous carcinoma cells. We show that, following TPT-PJ34 combined treatment, MCF7 cells exhibit apoptotic death while MDA-MB231 and SCC022 cells are more resistant to these agents. Specifically, in MCF7, (i) PJ34 in combination with TPT causes a G2/M cell cycle arrest followed by massive apoptosis; (ii) PJ34 addition reverts TPT-dependent PARP-1 automodification and triggers caspase-dependent PARP-1 proteolysis; (iii) TPT, used as a single agent, stimulates p53 expression while in combination with PJ34 increases p53, TAp63α and TAp63γ protein levels with a concomitant reduction of MDM2 protein. The identification of p63 proteins as new players involved in the cancer cell response to TPT-PJ34 is relevant for a better understanding of the PARP1-dependent signaling of DNA damage. Furthermore, our data indicate that, in response to TPT-PJ34 combined chemotherapy, a functional cooperation between p53 and TAp63 proteins may occur and be essential to trigger apoptotic cell death.
Collapse
|
99
|
Bayraktar S, Glück S. Molecularly targeted therapies for metastatic triple-negative breast cancer. Breast Cancer Res Treat 2013; 138:21-35. [PMID: 23358903 DOI: 10.1007/s10549-013-2421-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Accepted: 01/15/2013] [Indexed: 12/11/2022]
Abstract
Triple-negative breast cancer (TNBC) refers to a heterogeneous group of tumors that do not express the estrogen/progesterone-receptor (ER/PR), and human epidermal growth factor receptor-2 (HER2). TNBC is an aggressive histological subtype with limited treatment options and very poor prognosis following progression after standard chemotherapy regimens. There have been significant improvements in the outcome of other subtypes of breast cancer, including ER-positive/HER2 overexpressed tumors, attributed to the addition of targeted therapy, including hormonal agents and trastuzumab. However, no specific targeted agents are currently available for the treatment of TNBC. This review aims to collate and describe the most recent data on targeted therapies that have demonstrated efficacy in the management of metastatic TNBC. Targeted agents that have been investigated in the treatment of metastatic TNBC include inhibitors of poly(ADP-ribose) polymerase, angiogenesis, mammalian target of rapamycin, epidermal growth factor receptor, HDAC, Jak2, and Src. Several of these agents have shown considerable promise.
Collapse
Affiliation(s)
- Soley Bayraktar
- Department of Medical Oncology, Mercy Cancer Center, Ardmore, OK, USA.
| | | |
Collapse
|
100
|
Role of poly(ADP-ribose) polymerase-1 in the removal of UV-induced DNA lesions by nucleotide excision repair. Proc Natl Acad Sci U S A 2013; 110:1658-63. [PMID: 23319653 DOI: 10.1073/pnas.1209507110] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Among the earliest responses of mammalian cells to DNA damage is catalytic activation of a nuclear enzyme poly(ADP-ribose) polymerase-1 (PARP-1). Activated PARP-1 forms the polymers of ADP-ribose (pADPr or PAR) that posttranslationally modify its target proteins, such as PARP-1 and DNA repair-related proteins. Although this metabolism is known to be implicated in other repair pathways, here we show its role in the versatile nucleotide excision repair pathway (NER) that removes a variety of DNA damages including those induced by UV. We show that PARP inhibition or specific depletion of PARP-1 decreases the efficiency of removal of UV-induced DNA damage from human skin fibroblasts or mouse epidermis. Using NER-proficient and -deficient cells and in vitro PARP-1 assays, we show that damaged DNA-binding protein 2 (DDB2), a key lesion recognition protein of the global genomic subpathway of NER (GG-NER), associates with PARP-1 in the vicinity of UV-damaged chromatin, stimulates its catalytic activity, and is modified by pADPr. PARP inhibition abolishes UV-induced interaction of DDB2 with PARP-1 or xeroderma pigmentosum group C (XPC) and also decreases localization of XPC to UV-damaged DNA, which is a key step that leads to downstream events in GG-NER. Thus, PARP-1 collaborates with DDB2 to increase the efficiency of the lesion recognition step of GG-NER.
Collapse
|