1
|
Chahat, Nainwal N, Murti Y, Yadav S, Rawat P, Dhiman S, Kumar B. Advancements in targeting tumor suppressor genes (p53 and BRCA 1/2) in breast cancer therapy. Mol Divers 2025; 29:2691-2716. [PMID: 39152355 DOI: 10.1007/s11030-024-10964-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
Globally, among numerous cancer subtypes, breast cancer (BC) is one of the most prevalent forms of cancer affecting the female population. A female's family history significantly increases her risk of developing breast cancer. BC is caused by aberrant breast cells that proliferate and develop into tumors. It is estimated that 5-10% of breast carcinomas are inherited and involve genetic mutations that ensure the survival and prognosis of breast cancer cells. The most common genetic variations are responsible for hereditary breast cancer but are not limited to p53, BRCA1, and BRCA2. BRCA1 and BRCA2 are involved in genomic recombination, cell cycle monitoring, programmed cell death, and transcriptional regulation. When BRCA1 and 2 genetic variations are present in breast carcinoma, p53 irregularities become more prevalent. Both BRCA1/2 and p53 genes are involved in cell cycle monitoring. The present article discusses the current status of breast cancer research, spotlighting the tumor suppressor genes (BRCA1/2 and p53) along with structural activity relationship studies, FDA-approved drugs, and several therapy modalities for treating BC. Breast cancer drugs, accessible today in the market, have different side effects including anemia, pneumonitis, nausea, lethargy, and vomiting. Thus, the development of novel p53 and BRCA1/2 inhibitors with minimal possible side effects is crucial. We have covered compounds that have been examined subsequently (2020 onwards) in this overview which may be utilized as lead compounds. Further, we have covered mechanistic pathways to showcase the critical druggable targets and clinical and post-clinical drugs targeting them for their utility in BC.
Collapse
Affiliation(s)
- Chahat
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Srinagar, 246174, Uttarakhand, India
| | - Nidhi Nainwal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Premanagar, Dehradun, 248007, Uttarakhand, India
| | - Yogesh Murti
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| | - Savita Yadav
- IES Institute of Technology and Management, IES University, Bhopal, 462044, Madhya Pradesh, India
| | - Pramod Rawat
- Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
- Graphic Era Hill University Clement Town, Dehradun, 248002, India
| | - Sonia Dhiman
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Bhupinder Kumar
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Srinagar, 246174, Uttarakhand, India.
| |
Collapse
|
2
|
Vasigh M, Mohamed A, Jacobs L, Lange J, Camp M, Sun B, Wright P, O'Donnell M, Tran HT, Sogunro O, Habibi M, Johnston F, Euhus D. The Association Between Breast Cancer Predisposing Genetic Variants and Multifocal, Multicentric Breast Cancer. Ann Surg Oncol 2024; 31:8891-8899. [PMID: 39331289 DOI: 10.1245/s10434-024-16243-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/01/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Breast-conserving surgery is often discouraged in BRCA gene carriers with early onset breast cancer. The genetic variant carrier breast cancers are more likely to be multifocal or multicentric (MFMC). PATIENTS AND METHOD This retrospective study includes newly diagnosed patients with breast cancer undergoing genetic testing between 2010 and 2021 within the Johns Hopkins Regional Health System. After excluding patients who received neoadjuvant chemotherapy or stage IV breast cancers, patients were divided into two groups: those who tested positive for a variant recognized by the National Comprehensive Cancer Network as predisposing the patient to breast cancer (ATM, BRCA1, BRCA2, CHEK2, NF1, PALB2, RAD51C, RAD51D, and TP53) and those who tested negative. Pathologic features of the tumors were compared, focusing on evidence for MFMC disease, defined as more than one malignant foci more than 5 mm apart. RESULTS Among the 282 eligible cases, 69 (24%) were positive for a genetic variant. The variant carriers were younger at diagnosis (p < 0.001), more likely to have invasive ductal carcinoma (p = 0.03), more likely to have undergone mastectomy (p = 0.03), and more likely to have a grade 3 cancer (p = 0.003). Variant carriers were not more likely to have MFMC disease (28% vs. 22%, p = 0.4). A positive genetic variant was not a predictor of MFMC within the entire cohort [odds ratio (OR):1.3, 95% confidence interval (CI) 0.6-2.6, p = 0.5). CONCLUSION Genetic variant carrier cancers are not more likely to be MCMF than sporadic cancers.
Collapse
Affiliation(s)
- Mahtab Vasigh
- Department of Surgical Oncology, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - Ahmed Mohamed
- Department of Surgical Oncology, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - Lisa Jacobs
- Department of Surgical Oncology, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - Julie Lange
- Department of Surgical Oncology, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - Melissa Camp
- Department of Surgical Oncology, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - Bonnie Sun
- Department of Surgical Oncology, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - Pamela Wright
- Department of Surgical Oncology, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - Maureen O'Donnell
- Department of Surgical Oncology, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - Hanh-Tam Tran
- Department of Surgical Oncology, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - Olutayo Sogunro
- Department of Surgical Oncology, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - Mehran Habibi
- Department of Surgical Oncology, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - Fabian Johnston
- Department of Surgical Oncology, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - David Euhus
- Department of Surgical Oncology, Johns Hopkins Medical Institute, Baltimore, MD, USA.
| |
Collapse
|
3
|
Choi E, Mun GI, Lee J, Lee H, Cho J, Lee YS. BRCA1 deficiency in triple-negative breast cancer: Protein stability as a basis for therapy. Biomed Pharmacother 2023; 158:114090. [PMID: 36493696 DOI: 10.1016/j.biopha.2022.114090] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/24/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Mutations in breast cancer-associated 1 (BRCA1) increase the lifetime risk of developing breast cancer by up to 51% over the risk of the general population. Many aspects of this multifunctional protein have been revealed, including its essential role in homologous recombination repair, E3 ubiquitin ligase activity, transcriptional regulation, and apoptosis. Although most studies have focused on BRCA1 deficiency due to mutations, only a minority of patients carry BRCA1 mutations. A recent study has suggested an expanded definition of BRCA1 deficiency with reduced BRCA1 levels, which accounts for almost half of all triple-negative breast cancer (TNBC) patients. Reduced BRCA1 levels can result from epigenetic modifications or increased proteasomal degradation. In this review, we discuss how this knowledge of BRCA1 function and regulation of BRCA1 protein stability can help overcome the challenges encountered in the clinic and advance current treatment strategies for BRCA1-related breast cancer patients, especially focusing on TNBC.
Collapse
Affiliation(s)
- Eun Choi
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Gil-Im Mun
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Joohyun Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hanhee Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jaeho Cho
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Yun-Sil Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
4
|
Alsherbiny MA, Bhuyan DJ, Radwan I, Chang D, Li CG. Metabolomic Identification of Anticancer Metabolites of Australian Propolis and Proteomic Elucidation of Its Synergistic Mechanisms with Doxorubicin in the MCF7 Cells. Int J Mol Sci 2021; 22:ijms22157840. [PMID: 34360606 PMCID: PMC8346082 DOI: 10.3390/ijms22157840] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 12/11/2022] Open
Abstract
The combination of natural products with standard chemotherapeutic agents offers a promising strategy to enhance the efficacy or reduce the side effects of standard chemotherapy. Doxorubicin (DOX), a standard drug for breast cancer, has several disadvantages, including severe side effects and the development of drug resistance. Recently, we reported the potential bioactive markers of Australian propolis extract (AP-1) and their broad spectrum of pharmacological activities. In the present study, we explored the synergistic interactions between AP-1 and DOX in the MCF7 breast adenocarcinoma cells using different synergy quantitation models. Biochemometric and metabolomics-driven analysis was performed to identify the potential anticancer metabolites in AP-1. The molecular mechanisms of synergy were studied by analysing the apoptotic profile via flow cytometry, apoptotic proteome array and measuring the oxidative status of the MCF7 cells treated with the most synergistic combination. Furthermore, label-free quantification proteomics analysis was performed to decipher the underlying synergistic mechanisms. Five prenylated stilbenes were identified as the key metabolites in the most active AP-1 fraction. Strong synergy was observed when AP-1 was combined with DOX in the ratio of 100:0.29 (w/w) as validated by different synergy quantitation models implemented. AP-1 significantly enhanced the inhibitory effect of DOX against MCF7 cell proliferation in a dose-dependent manner with significant inhibition of the reactive oxygen species (p < 0.0001) compared to DOX alone. AP-1 enabled the reversal of DOX-mediated necrosis to programmed cell death, which may be advantageous to decline DOX-related side effects. AP-1 also significantly enhanced the apoptotic effect of DOX after 24 h of treatment with significant upregulation of catalase, HTRA2/Omi, FADD together with DR5 and DR4 TRAIL-mediated apoptosis (p < 0.05), contributing to the antiproliferative activity of AP-1. Significant upregulation of pro-apoptotic p27, PON2 and catalase with downregulated anti-apoptotic XIAP, HSP60 and HIF-1α, and increased antioxidant proteins (catalase and PON2) may be associated with the improved apoptosis and oxidative status of the synergistic combination-treated MCF7 cells compared to the mono treatments. Shotgun proteomics identified 21 significantly dysregulated proteins in the synergistic combination-treated cells versus the mono treatments. These proteins were involved in the TP53/ATM-regulated non-homologous end-joining pathway and double-strand breaks repairs, recruiting the overexpressed BRCA1 and suppressed RIF1 encoded proteins. The overexpression of UPF2 was noticed in the synergistic combination treatment, which could assist in overcoming doxorubicin resistance-associated long non-coding RNA and metastasis of the MCF7 cells. In conclusion, we identified the significant synergy and highlighted the key molecular pathways in the interaction between AP-1 and DOX in the MCF7 cells together with the AP-1 anticancer metabolites. Further in vivo and clinical studies are warranted on this synergistic combination.
Collapse
Affiliation(s)
- Muhammad A. Alsherbiny
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
- Correspondence: (M.A.A.); (D.J.B.); (C.-G.L.)
| | - Deep J. Bhuyan
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
- Correspondence: (M.A.A.); (D.J.B.); (C.-G.L.)
| | - Ibrahim Radwan
- Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia;
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
| | - Chun-Guang Li
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
- Correspondence: (M.A.A.); (D.J.B.); (C.-G.L.)
| |
Collapse
|
5
|
Lee A, Moon BI, Kim TH. BRCA1/ BRCA2 Pathogenic Variant Breast Cancer: Treatment and Prevention Strategies. Ann Lab Med 2020; 40:114-121. [PMID: 31650727 PMCID: PMC6822003 DOI: 10.3343/alm.2020.40.2.114] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/21/2019] [Accepted: 10/10/2019] [Indexed: 12/24/2022] Open
Abstract
Hereditary breast cancer is known for its strong tendency of inheritance. Most hereditary breast cancers are related to BRCA1/BRCA2 pathogenic variants. The lifelong risk of breast cancer in pathogenic BRCA1 and BRCA2 variant carriers is approximately 65% and 45%, respectively, whereas that of ovarian cancer is estimated to be 39% and 11%, respectively. Therefore, understanding these variants and clinical knowledge on their occurrence in breast cancers and carriers are important. BRCA1 pathogenic variant breast cancer shows more aggressive clinicopathological features than the BRCA2 pathogenic variant breast cancer. Compared with sporadic breast cancer, their prognosis is still debated. Treatments of BRCA1/BRCA2 pathogenic variant breast cancer are similar to those for BRCA-negative breast cancer, mainly including surgery, radiotherapy, and chemotherapy. Recently, various clinical trials have investigated poly (adenosine diphosphate [ADP]-ribose) polymerase (PARP) inhibitor treatment for advanced-stage BRCA1/BRCA2 pathogenic variant breast cancer. Among the various PARP inhibitors, olaparib and talazoparib, which reached phase III clinical trials, showed improvement of median progression-free survival around three months. Preventive and surveillance strategies for BRCA pathogenic variant breast cancer to reduce cancer recurrence and improve treatment outcomes have recently received increasing attention. In this review, we provide an information on the clinical features of BRCA1/BRCA2 pathogenic variant breast cancer and clinical recommendations for BRCA pathogenic variant carriers, with a focus on treatment and prevention strategies. With this knowledge, clinicians could manage the BRCA1/BRCA2 pathogenic variant breast cancer patients more effectively.
Collapse
Affiliation(s)
- Anbok Lee
- Department of Surgery, Busan Paik Hospital, Inje University College of Medicine, Busan, Korea.
| | - Byung In Moon
- Department of Surgery, Mokdong Hospital, Ewha Womans University College of Medicine, Seoul, Korea
| | - Tae Hyun Kim
- Department of Surgery, Busan Paik Hospital, Inje University College of Medicine, Busan, Korea
| |
Collapse
|
6
|
Wang T, McCullough LE, White AJ, Bradshaw PT, Xu X, Cho YH, Terry MB, Teitelbaum SL, Neugut AI, Santella RM, Chen J, Gammon MD. Prediagnosis aspirin use, DNA methylation, and mortality after breast cancer: A population-based study. Cancer 2019; 125:3836-3844. [PMID: 31402456 DOI: 10.1002/cncr.32364] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 12/20/2018] [Accepted: 01/07/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND The authors hypothesized that epigenetic changes may help to clarify the underlying biologic mechanism linking aspirin use to breast cancer prognosis. To the authors' knowledge, this is the first epidemiologic study to examine whether global methylation and/or tumor promoter methylation of breast cancer-related genes interact with aspirin use to impact mortality after breast cancer. METHODS Prediagnosis aspirin use was assessed through in-person interviews within a population-based cohort of 1508 women diagnosed with a first primary breast cancer in 1996 and 1997. Global methylation in peripheral blood was assessed by long interspersed elements-1 (LINE-1) and the luminometric methylation assay. Promoter methylation of 13 breast cancer-related genes was measured in tumor by methylation-specific polymerase chain reaction and the MethyLight assay. Vital status was determined by the National Death Index through December 31, 2014 (N = 202/476 breast cancer-specific/all-cause deaths identified among 1266 women with any methylation assessment and complete aspirin data). Cox proportional hazards regression was used to estimate hazard ratios (HRs) and 95% CIs, and the likelihood ratio test was used to evaluate multiplicative interactions. RESULTS All-cause mortality was elevated among aspirin users who had methylated promotor of BRCA1 (HR, 1.67; 95% CI, 1.26-2.22), but not among those with unmethylated promoter of BRCA1 (HR, 0.99; 95% CI, 0.67-1.45; P for interaction ≤.05). Decreased breast cancer-specific mortality was observed among aspirin users who had unmethylated promotor of BRCA1 and PR and global hypermethylation of LINE-1 (HR, 0.60, 0.78, and 0.63, respectively; P for interaction ≤.05), although the 95% CIs included the null. CONCLUSIONS The current study suggests that the LINE-1 global methylation and promoter methylation of BRCA1 and PR in tumor may interact with aspirin use to influence mortality after breast cancer.
Collapse
Affiliation(s)
- Tengteng Wang
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | | | - Alexandra J White
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Patrick T Bradshaw
- Division of Epidemiology, University of California, Berkeley, California
| | - Xinran Xu
- Department of Biometrics, Roche Product Development in Asia-Pacific, Shanghai, China
| | - Yoon Hee Cho
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana
| | - Mary Beth Terry
- Department of Epidemiology, Columbia University, New York, New York
| | - Susan L Teitelbaum
- Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alfred I Neugut
- Department of Epidemiology, Columbia University, New York, New York.,Department of Medicine, Columbia University, New York, New York
| | | | - Jia Chen
- Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marilie D Gammon
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
7
|
Zhu Y, Zhai K, Ke J, Li J, Gong Y, Yang Y, Tian J, Zhang Y, Zou D, Peng X, Gong J, Zhong R, Huang K, Chang J, Miao X. BRCA1 missense polymorphisms are associated with poor prognosis of pancreatic cancer patients in a Chinese population. Oncotarget 2018; 8:36033-36039. [PMID: 28415599 PMCID: PMC5482636 DOI: 10.18632/oncotarget.16422] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/15/2017] [Indexed: 01/24/2023] Open
Abstract
Pancreatic cancer is a highly lethal disease with limited prognostic marker. BRAC1 and BRCA2 are two classic tumor suppressor genes which play an important role in DNA repair. Somatic mutations and germline genetic variants on BRCA1/2 have been found associated with the tumorigenesis of pancreatic cancer. However, the correlations between BRCA1/2 polymorphism and pancreatic cancer prognosis remained unknown. In this study, we genotyped three tag missense variants on BRCA1/2 in 603 sporadic pancreatic cancer patients in a Chinese population. We found rs1799966 on BRCA1 was associated with poor prognosis of pancreatic cancer patients with hazard ratio being 1.23 (95% CI: 1.09–1.40, P = 0.0010). Further stratification analyses showed that significant correlation was particularly in locally advanced stage patients with hazard ratio being 1.36 (95% CI: 1.13–1.64, P = 0.0014), but not in patients in local stage (P = 0.1139) or metastatic stage (P = 0.5185). Two missense variants (rs766173 and rs144848) on BRAC2 showed no significant correlation with pancreatic cancer patients’ overall survival. In conclusion, we identified a germline missense variant on BRAC1 significantly associated with poor prognosis of pancreatic cancer patients with locally advanced stage. These results may contribute to the precision medicine of this disease.
Collapse
Affiliation(s)
- Ying Zhu
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kan Zhai
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Juntao Ke
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaoyuan Li
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yajie Gong
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Yang
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianbo Tian
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Zhang
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danyi Zou
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiating Peng
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Gong
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Zhong
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Huang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Jiang Chang
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Miao
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Donaire JM, Peralta O, Bravo ME. MANEJO QUIRÚRGICO DE LA PACIENTE CON CÁNCER DE MAMA PORTADORA DE MUTACIONES GENÉTICAS. REVISTA MÉDICA CLÍNICA LAS CONDES 2017. [DOI: 10.1016/j.rmclc.2017.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
9
|
BRCA mutation genetic testing implications in the United States. Breast 2016; 31:224-232. [PMID: 27931006 DOI: 10.1016/j.breast.2016.11.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 12/14/2022] Open
Abstract
BRCA mutation carriers have a very high risk of breast and ovarian cancer by age 70, in the ranges 47%-66% and 40%-57%, respectively. Additionally, women with BRCA mutation-associated breast cancer also have an elevated risk of other or secondary malignancies. Fortunately, the breast and ovarian cancer outcome for BRCA1/2 mutation carriers is at least as good as for non-carriers with chemoprevention, prophylactic surgeries and appropriate use of therapies. Therefore, identification of those who might have a mutation is important so that genetic counseling, testing, screening and prevention strategies can be applied in a timely manner. This article reviews the impact of genetic testing in general, timing of genetic testing after diagnosis and prior knowledge of mutation status in BRCA carriers with newly diagnosed breast cancer. Additionally, risk-reducing surgeries including the prophylactic contralateral mastectomy, and bilateral salpingo-oophorectomy and the sensitivity of BRCA-defective breast cancer cell lines to differential chemotherapeutic agents will be discussed.
Collapse
|
10
|
New Insights Into the Mechanism of COP9 Signalosome-Cullin-RING Ubiquitin-Ligase Pathway Deregulation in Urological Cancers. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 323:181-229. [PMID: 26944622 DOI: 10.1016/bs.ircmb.2015.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Urological cancers are a very common type of cancer worldwide and have alarming high incidence and mortality rates, especially in kidney cancers, illustrate the urgent need for new therapeutic targets. Recent publications point to a deregulated COP9 signalosome (CSN)-cullin-RING ubiquitin-ligase (CRL) pathway which is here considered and investigated as potential target in urological cancers with strong focus on renal cell carcinomas (RCC). The CSN forms supercomplexes with CRLs in order to preserve protein homeostasis and was found deregulated in several cancer types. Examination of selected CSN-CRL pathway components in RCC patient samples and four RCC cell lines revealed an interesting deregulated p27(Kip1)-Skp2-CAND1 axis and two p27(Kip1) point mutations in 786-O cells; p27(Kip1)V109G and p27(Kip1)I119T. The p27(Kip1) mutants were detected in patients with RCC and appear to be responsible for an accelerated growth rate in 786-O cells. The occurrence of p27(Kip1)V109G and p27(Kip1)I119T in RCC makes the CSN-CRL pathway an attractive therapeutic target.
Collapse
|
11
|
van den Broek AJ, van 't Veer LJ, Hooning MJ, Cornelissen S, Broeks A, Rutgers EJ, Smit VTHBM, Cornelisse CJ, van Beek M, Janssen-Heijnen ML, Seynaeve C, Westenend PJ, Jobsen JJ, Siesling S, Tollenaar RAEM, van Leeuwen FE, Schmidt MK. Impact of Age at Primary Breast Cancer on Contralateral Breast Cancer Risk in BRCA1/2 Mutation Carriers. J Clin Oncol 2015; 34:409-18. [PMID: 26700119 DOI: 10.1200/jco.2015.62.3942] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE To determine prospectively overall and age-specific estimates of contralateral breast cancer (CBC) risk for young patients with breast cancer with or without BRCA1/2 mutations. PATIENTS AND METHODS A cohort of 6,294 patients with invasive breast cancer diagnosed under 50 years of age and treated between 1970 and 2003 in 10 Dutch centers was tested for the most prevalent BRCA1/2 mutations. We report absolute risks and hazard ratios within the cohort from competing risk analyses. RESULTS After a median follow-up of 12.5 years, 578 CBCs were observed in our study population. CBC risk for BRCA1 and BRCA2 mutation carriers was two to three times higher than for noncarriers (hazard ratios, 3.31 [95% CI, 2.41 to 4.55; P < .001] and 2.17 [95% CI,1.22 to 3.85; P = .01], respectively). Ten-year cumulative CBC risks were 21.1% (95% CI, 15.4 to 27.4) for BRCA1, 10.8% (95% CI, 4.7 to 19.6) for BRCA2 mutation carriers and 5.1% (95% CI, 4.5 to 5.7) for noncarriers. Age at diagnosis of the first breast cancer was a significant predictor of CBC risk in BRCA1/2 mutation carriers only; those diagnosed before age 41 years had a 10-year cumulative CBC risk of 23.9% (BRCA1: 25.5%; BRCA2: 17.2%) compared with 12.6% (BRCA1: 15.6%; BRCA2: 7.2%) for those 41 to 49 years of age (P = .02); our review of published studies showed ranges of 24% to 31% before age 40 years (BRCA1: 24% to 32%; BRCA2:17% to 29%) and 8% to 21% after 40 years (BRCA1: 11% to 52%; BRCA2: 7% to 18%), respectively. CONCLUSION Age at first breast cancer is a strong risk factor for cumulative CBC risk in BRCA1/2 mutation carriers. Considering the available evidence, age-specific risk estimates should be included in counseling.
Collapse
Affiliation(s)
- Alexandra J van den Broek
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Laura J van 't Veer
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Maartje J Hooning
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Sten Cornelissen
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Annegien Broeks
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Emiel J Rutgers
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Vincent T H B M Smit
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Cees J Cornelisse
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Mike van Beek
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Maryska L Janssen-Heijnen
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Caroline Seynaeve
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Pieter J Westenend
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Jan J Jobsen
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Sabine Siesling
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Rob A E M Tollenaar
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Flora E van Leeuwen
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands
| | - Marjanka K Schmidt
- Alexandra J. van den Broek, Laura J. van 't Veer, Sten Cornelissen, Annegien Broeks, Emiel J. Rutgers, Flora E. van Leeuwen, and Marjanka K. Schmidt, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Maartje J. Hooning and Caroline Seynaeve, Erasmus MC Cancer Institute, Rotterdam; Vincent T.H.B.M. Smit, Cees J. Cornelisse, and Rob A.E.M. Tollenaar, Leiden University Medical Center, Leiden; Mike van Beek, PAMM, Catharina Hospital, Eindhoven; Maryska L. Janssen-Heijnen, Netherlands Comprehensive Cancer Organization, Eindhoven, and VieCuri Medical Centre, Venlo; Pieter J. Westenend, Laboratory for Pathology, Dordrecht; Jan J. Jobsen, Medisch Spectrum Twente, Enschede; and Sabine Sieling, Netherlands Comprehensive Cancer Organization, Utrecht, Netherlands.
| |
Collapse
|
12
|
van den Broek AJ, Schmidt MK, van 't Veer LJ, Tollenaar RAEM, van Leeuwen FE. Worse breast cancer prognosis of BRCA1/BRCA2 mutation carriers: what's the evidence? A systematic review with meta-analysis. PLoS One 2015; 10:e0120189. [PMID: 25816289 PMCID: PMC4376645 DOI: 10.1371/journal.pone.0120189] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 01/26/2015] [Indexed: 01/16/2023] Open
Abstract
Objective Conflicting conclusions have been published regarding breast cancer survival of BRCA1/2 mutation carriers. Here we provide an evidence-based systematic literature review. Methods Eligible publications were observational studies assessing the survival of breast cancer patients carrying a BRCA1/2 mutation compared to non-carriers or the general breast cancer population. We performed meta-analyses and best-evidence syntheses for survival outcomes taking into account study quality assessed by selection bias, misclassification bias and confounding. Results Sixty-six relevant studies were identified. Moderate evidence for a worse unadjusted recurrence-free survival for BRCA1 mutation carriers was found. For BRCA1 and BRCA2 there was a tendency towards a worse breast cancer-specific and overall survival, however, results were heterogeneous and the evidence was judged to be indecisive. Surprisingly, only 8 studies considered adjuvant treatment as a confounder or effect modifier while only two studies took prophylactic surgery into account. Adjustment for tumour characteristics tended to shift the observed risk estimates towards a relatively more favourable survival. Conclusions In contrast to currently held beliefs of some oncologists, current evidence does not support worse breast cancer survival of BRCA1/2 mutation carriers in the adjuvant setting; differences if any are likely to be small. More well-designed studies are awaited.
Collapse
Affiliation(s)
- Alexandra J van den Broek
- Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Marjanka K Schmidt
- Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, Netherlands; Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Laura J van 't Veer
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Rob A E M Tollenaar
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - Flora E van Leeuwen
- Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
13
|
Molina-Montes E, Pérez-Nevot B, Pollán M, Sánchez-Cantalejo E, Espín J, Sánchez MJ. Cumulative risk of second primary contralateral breast cancer in BRCA1/BRCA2 mutation carriers with a first breast cancer: A systematic review and meta-analysis. Breast 2014; 23:721-42. [PMID: 25467311 DOI: 10.1016/j.breast.2014.10.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 10/05/2014] [Accepted: 10/12/2014] [Indexed: 12/19/2022] Open
Abstract
BRCA1/2 mutation carriers are at a higher risk of breast cancer and of subsequent contralateral breast cancer (CBC). This study aims to evaluate the evidence of the effect of the BRCA1/2-carriership on CBC cumulative risk in female breast cancer patients. The literature was searched in Pubmed and Embase up to June 2013 for studies on CBC risk after a first primary invasive breast cancer in female BRCA1/2 mutation carriers. A qualitative synthesis was carried out and the methodological quality of the studies evaluated. Cumulative risks of CBC after 5, 10 and 15 years since the first breast cancer diagnosis were pooled by BRCA1/2 mutation status. A total number of 20 articles, out of 1324 retrieved through the search, met the inclusion criteria: 18 retrospective and 2 prospective cohort studies. Cumulative risks of up to five studies were pooled. The cumulative 5-years risk of CBC for BRCA1 and BRCA2 mutation carriers was 15% (95% CI: 9.5%-20%) and 9% (95% CI: 5%-14%), respectively. This risk increases with time since diagnosis of the first breast cancer; the 10-years risk increased up to 27% and 19%, respectively. The 5-years cumulative risk was remarkably lower in non-BRCA carriers (3%; 95% CI: 2%-5%) and remained so over subsequent years (5%; 95% CI: 3%-7%). In conclusion, risk of CBC increases with length of time after the first breast cancer diagnosis in BRCA1/2 mutation carriers. Studies addressing the impact of treatment-related factors and clinical characteristics of the first breast cancer on this risk are warranted.
Collapse
Affiliation(s)
- Esther Molina-Montes
- Granada Cancer Registry, Andalusian School of Public Health, Granada, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Spain; Instituto de Investigación Biosanitaria de Granada (ibs.Granada), Hospitales Universitarios de Granada, Universidad de Granada, Granada, Spain
| | - Beatriz Pérez-Nevot
- Clinical Analysis Service, Virgen de la Victoria University Hospital, Malaga, Spain
| | - Marina Pollán
- CIBER de Epidemiología y Salud Pública (CIBERESP), Spain; Cancer Epidemiology Unit, National Center for Epidemiology, Instituto de Salud Carlos III, Madrid, Spain; Cancer Epidemiology Research Group, Oncology and Hematology Area, IIS Puerta de Hierro (IDIPHIM), Madrid, Spain
| | - Emilio Sánchez-Cantalejo
- Granada Cancer Registry, Andalusian School of Public Health, Granada, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Spain; Instituto de Investigación Biosanitaria de Granada (ibs.Granada), Hospitales Universitarios de Granada, Universidad de Granada, Granada, Spain
| | - Jaime Espín
- Granada Cancer Registry, Andalusian School of Public Health, Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.Granada), Hospitales Universitarios de Granada, Universidad de Granada, Granada, Spain
| | - María-José Sánchez
- Granada Cancer Registry, Andalusian School of Public Health, Granada, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Spain; Instituto de Investigación Biosanitaria de Granada (ibs.Granada), Hospitales Universitarios de Granada, Universidad de Granada, Granada, Spain.
| |
Collapse
|
14
|
Surgical management of breast cancer in BRCA-mutation carriers: a systematic review and meta-analysis. Breast Cancer Res Treat 2014; 144:443-55. [PMID: 24567198 DOI: 10.1007/s10549-014-2890-1] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 02/17/2014] [Indexed: 02/04/2023]
Abstract
This meta-analysis investigates the oncological safety of breast-conserving therapy BCT in BRCA-mutation carriers and the risk for contralateral breast cancer (CBC) compared with non-carriers, potential risk factors for ipsilateral breast recurrence (IBR) or CBC and grades these factors based on the level of evidence. A PubMed search was conducted through April 2013 to identify studies that described the risk for IBR and CBC after BCT in BRCA-mutation carriers versus non-carriers as well as studies that investigated risk factors for IBR and CBC in BRCA-mutation carriers. Results were summarized using meta-analysis when sufficient studies were available. Ten studies investigated the oncological safety of BCT in BRCA-mutation carriers versus non-carriers. There was no significant difference in IBR between carriers and controls (RR 1.45, 95 % CI 0.98-2.14). However, a significant higher risk for IBR in BRCA-mutation carriers was observed in studies with a median follow-up ≥7 years (RR 1.51, 95 % CI 1.15-1.98). CBCs were significantly greater in carriers versus controls (RR 3.56, 95 % CI 2.50-5.08). Use of adjuvant chemotherapy and oophorectomy were associated with a significantly lower risk for IBR in BRCA-mutation carriers. Three factors were associated with a lower risk for CBC in BRCA-mutation carriers: oophorectomy, use of tamoxifen, and age at first breast cancer. Based on current evidence, the use of BCT in BRCA-mutation carriers can be considered a reasonable option since it does not seem to increase the risk for IBR. However, several aspects should be taken into account before the final decision-making.
Collapse
|
15
|
Dressler AC, Hudelist G, Fink-Retter A, Gschwantler-Kaulich D, Pfeiler G, Rosner M, Hengstschläger M, Singer CF. Tuberin and p27 expression in breast cancer patients with or without BRCA germline mutations. J Cancer Res Clin Oncol 2013; 139:1349-55. [PMID: 23689538 DOI: 10.1007/s00432-013-1443-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 04/16/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND Tuberin, the protein product of tuberous sclerosis gene 2 (TSC2), is the functional component of the TSC1/TSC2 complex and regulates cell cycle through activation of the cyclin-dependent kinase inhibitor p27. The transcriptional regulation of p27 is, however, also linked to a functional BRCA protein, since in BRCA1 mutant breast cancer cells, which lack the ability to repair DNA damages by homologous recombination, p27 is down-regulated. We have therefore investigated the expression of both tuberin and p27 in normal breast tissue, and in malignant epithelium from women with and without a BRCA mutation. MATERIALS AND METHODS immunohistochemistry was used to compare p27 and tuberin protein expression in 26 BRCA1 and 2 mutation carriers, in 53 matched breast cancer patients without a family history, and in 74 benign breast tissues in a case-control study. RESULTS Tuberin and p27 protein expression were significantly more common in benign when compared to malignant breast tissue (p = 0.01 and p = 0.03), but no difference was observed when sporadic and BRCA-mutated breast cancer specimen were compared. Tuberin and p27 were positively correlated with each other (p = 0.0017, r = 0.2527). Furthermore, p27 expression was positively correlated with ER and PR, and negatively correlated with tumor size. The expression of tuberin and p27 in breast cancer was not correlated with clinical outcome. CONCLUSION Our results suggest that tuberin and p27 are aberrantly expressed in malignant tissue, but their expression does not appear to be dependent on the BRCA mutation state of a breast cancer patient.
Collapse
Affiliation(s)
- Anne Catharina Dressler
- Clinical Division of Gynaecology and Gynaecological Oncology, Department of Obstretrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Wu ZB, Cai L, Lin SJ, Xiong ZK, Lu JL, Mao Y, Yao Y, Zhou LF. High-mobility group box 2 is associated with prognosis of glioblastoma by promoting cell viability, invasion, and chemotherapeutic resistance. Neuro Oncol 2013; 15:1264-75. [PMID: 23828241 DOI: 10.1093/neuonc/not078] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The expression profile of high-mobility group box 2 (HMGB2) in patients with glioblastoma multiforme (GBM) and its clinical signature with underlying mechanisms were not fully explored. METHODS HMGB2 protein levels were measured in 51 GBM patients by immunohistochemical studies. To clarify the precise role of HMGB2 on cell invasion and viability of 3 GBM cell lines, we did in vitro and in vivo analyses with lentivirus vectors and small interfering RNA. Transwell invasion assays and wound-healing assays were used to analyze the invasion of GBM cells. Expression of p53 and matrix metalloproteinase 2/tissue inhibitors of metalloproteinase 2 (MMP2/TIMP2) protein was analyzed by Western blot. RESULTS HMGB2 protein expression was significantly higher in GBM than in controlled brain tissues (P < .0001). HMGB2 overexpression was significantly correlated with shorter overall survival time, which was the only independent prognostic factor for overall survival in a multivariate analysis (P = .017). HMGB2 knockdown by small interfering RNA decreased cell viability and invasion in vitro and significantly decreased tumor volume in vivo, which might be involved in the change of p53 expression and the balance of MMP2/TIMP2. Moreover, silencing of HMGB2 could significantly increase the sensitivity of GBM cells to temozolomide chemotherapy. CONCLUSIONS Our present data suggest that HMGB2 expression is a significant prognostic factor and might play an important role in cell invasion and temozolomide-induced chemotherapeutic sensitivity of GBM. This study highlights the importance of HMGB2 as a novel prognostic marker and an attractive therapeutic target of GBM.
Collapse
Affiliation(s)
- Zhe Bao Wu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Stecklein SR, Jensen RA. Identifying and exploiting defects in the Fanconi anemia/BRCA pathway in oncology. Transl Res 2012; 160:178-97. [PMID: 22683426 DOI: 10.1016/j.trsl.2012.01.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 01/18/2012] [Accepted: 01/19/2012] [Indexed: 01/07/2023]
Abstract
Defects in components of DNA repair pathways are responsible for numerous hereditary cancer syndromes and are also common in many sporadic malignancies. Inherited mutations in the breast cancer susceptibility genes BRCA1 and BRCA2 or components of the Fanconi anemia (FA) complex incite genomic instability and predispose to malignancy. The products of the BRCA and FA genes participate in a conserved DNA damage repair pathway that is responsible for repairing interstrand crosslinks and double-strand DNA breaks by homologous recombination. While the genetic instability resulting from FA/BRCA dysfunction contributes to cancer pathogenesis, deficiency of these genes also lends to therapeutic exploitation. Crosslinking agents and ionizing radiation induce damage in cancer cells that requires the FA/BRCA pathway to be resolved; thus cancers that are deficient in BRCA1, BRCA2, or any other component of the FA/BRCA pathway are hypersensitive to these agents. Moreover, emerging synthetic lethal strategies offer opportunities to selectively target cancer cells with defects in homologous recombination. Conversely, enhanced activity of the FA/BRCA pathway is responsible for acquired resistance to specific therapeutic agents, suggesting that both dysfunction and hyperfunction of the FA/BRCA repair machinery are rational targets for cancer therapy. Selection of specific cytotoxic agents based on repair capacity may improve responses and enable personalized cytotoxic chemotherapy. This article reviews the FA/BRCA pathway and current approaches to identify deficiencies within it, discusses synthetic lethality and enhanced repair capacity as causes of therapeutic hypersensitivity and resistance, respectively, and highlights recent studies that have linked FA/BRCA pathway function with therapeutic efficacy.
Collapse
Affiliation(s)
- Shane R Stecklein
- Department of Pathology and Laboratory Medicine and The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA
| | | |
Collapse
|
18
|
Shu XO, Long J, Lu W, Li C, Chen WY, Delahanty R, Cheng J, Cai H, Zheng Y, Shi J, Gu K, Wang WJ, Kraft P, Gao YT, Cai Q, Zheng W. Novel genetic markers of breast cancer survival identified by a genome-wide association study. Cancer Res 2012; 72:1182-9. [PMID: 22232737 DOI: 10.1158/0008-5472.can-11-2561] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Only two genome-wide association studies (GWAS) have been conducted to date to identify potential markers for total mortality after diagnosis of breast cancer. Here, we report the identification of two single-nucleotide polymorphisms (SNP) associated with total mortality from a two-stage GWAS conducted among 6,110 Shanghai-resident Chinese women with tumor-node-metastasis (TNM) stage I to IV breast cancer. The discovery stage included 1,950 patients and evaluated 613,031 common SNPs. The top 49 associations were evaluated in an independent replication stage of 4,160 Shanghai patients with breast cancer. A consistent and highly significant association with total mortality was documented for SNPs rs3784099 and rs9934948. SNP rs3784099, located in the RAD51L1 gene, was associated with total morality in both the discovery stage (P = 1.44 × 10(-8)) and replication stage (P = 0.06; P-combined = 1.17 × 10(-7)). Adjusted HRs for total mortality were 1.41 [95% confidence interval (CI), 1.18-1.68] for the AG genotype and 2.64 (95% CI, 1.74-4.03) for the AA genotype, when compared with the GG genotype. The variant C allele of rs9934948, located on chromosome 16, was associated with a similarly elevated risk of total mortality (P-combined = 5.75 × 10(-6)). We also observed this association among 1,145 patients with breast cancer of European ancestry from the Nurses' Health Study (NHS; P = 0.006); the association was highly significant in a combined analysis of NHS and Chinese data (P = 1.39 × 10(-7)). Similar associations were observed for these two SNPs with breast cancer-specific mortality. This study provides strong evidence suggesting that the RAD51L1 gene and a chromosome 16 locus influence breast cancer prognosis.
Collapse
Affiliation(s)
- Xiao Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37203, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Goodwin PJ, Phillips KA, West DW, Ennis M, Hopper JL, John EM, O'Malley FP, Milne RL, Andrulis IL, Friedlander ML, Southey MC, Apicella C, Giles GG, Longacre TA. Breast cancer prognosis in BRCA1 and BRCA2 mutation carriers: an International Prospective Breast Cancer Family Registry population-based cohort study. J Clin Oncol 2011; 30:19-26. [PMID: 22147742 DOI: 10.1200/jco.2010.33.0068] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To compare breast cancer prognosis in BRCA1 and BRCA2 mutation carriers with that in patients with sporadic disease. PATIENTS AND METHODS An international population-based cohort study was conducted in Canada, the United States, and Australia of 3,220 women with incident breast cancer diagnosed between 1995 and 2000 and observed prospectively. Ninety-three had BRCA1 mutations; 71, BRCA2 mutations; one, both mutations; 1,550, sporadic breast cancer; and 1,505, familial breast cancer (without known BRCA1 or BRCA2 mutation). Distant recurrence and death were analyzed. RESULTS Mean age at diagnosis was 45.3 years; mean follow-up was 7.9 years. Risks of distant recurrence and death did not differ significantly between BRCA1 mutation carriers and those with sporadic disease in univariable and multivariable analyses. Risk of distant recurrence was higher for BRCA2 mutation carriers compared with those with sporadic disease in univariable analysis (hazard ratio [HR], 1.63; 95% CI, 1.02 to 2.60; P = .04). Risk of death was also higher in BRCA2 carriers in univariable analysis (HR, 1.81; 95% CI, 1.15 to 2.86; P = .01). After adjustment for age, tumor stage and grade, nodal status, hormone receptors, and year of diagnosis, no differences were observed for distant recurrence (HR, 1.00; 95% CI, 0.62 to 1.61; P = 1.00) or death (HR, 1.12; 95% CI, 0.70 to 1.79; P = .64). CONCLUSION Outcomes of BRCA1 mutation carriers were similar to those of patients with sporadic breast cancer. Worse outcomes in BRCA2 mutation carriers in univariable analysis seem to reflect the presence of more adverse tumor characteristics in these carriers. Similar outcomes were identified in BRCA2 carriers and those with sporadic disease in multivariable analyses.
Collapse
Affiliation(s)
- Pamela J Goodwin
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Arun B, Bayraktar S, Liu DD, Gutierrez Barrera AM, Atchley D, Pusztai L, Litton JK, Valero V, Meric-Bernstam F, Hortobagyi GN, Albarracin C. Response to neoadjuvant systemic therapy for breast cancer in BRCA mutation carriers and noncarriers: a single-institution experience. J Clin Oncol 2011; 29:3739-46. [PMID: 21900106 PMCID: PMC4874218 DOI: 10.1200/jco.2011.35.2682] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
PURPOSE To compare the pathologic complete response (pCR) rate and relapse-free survival (RFS) and overall survival (OS) after neoadjuvant systemic chemotherapy (NST) in patients with breast cancer with and without deleterious BRCA1 and BRCA2 mutations. PATIENTS AND METHODS A total of 317 women who underwent BRCA genetic testing and were treated with NST for breast cancer between 1997 and 2009 were included in the study. The Kaplan-Meier product-limit method was used to estimate RFS and OS rates. Logistic regression models were fit to determine the associations between BRCA status, pCR, and survival. RESULTS Fifty-seven (18%) and 23 (7%) patients had BRCA1 and BRCA2 mutations, respectively. Twenty-six (46%) of 57 BRCA1 carriers achieved a pCR, compared with three (13%) of 23 BRCA2 carriers and 53 (22%) of 237 BRCA noncarriers (P < .001). In the multivariate logistic model, BRCA1 status (odds ratio [OR] = 3.16; 95% CI, 1.55 to 6.42; P = .002), estrogen receptor (ER) negativity (OR = 1.96; 95% CI:1.05 to 3.65; P = .03) and concurrent trastuzumab use (OR = 4.18; 95% CI, 2.04 to 8.57; P < .001) remained as independent significant predictors for a pCR. At a median follow-up of 3.2 years, 69 patients (22%) experienced a disease recurrence or death. No significant differences were noted in survival outcomes with respect to BRCA status and type of NST received. However, among BRCA1 carriers, patients who achieved a pCR had better 5-year RFS (P = .001) and OS (P = .01) rates than patients who did not. CONCLUSION BRCA1 status and ER negativity are independently associated with higher pCR rates in patients with breast cancer. Overall prognosis of breast cancer in BRCA carriers is similar to sporadic breast cancers.
Collapse
Affiliation(s)
- Banu Arun
- All authors: The University of Texas MD Anderson Cancer Center, Houston, TX.,Corresponding author: Banu Arun, MD, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1354, Houston, TX 77030-4009; e-mail:
| | - Soley Bayraktar
- All authors: The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Diane D. Liu
- All authors: The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Deann Atchley
- All authors: The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lajos Pusztai
- All authors: The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Vicente Valero
- All authors: The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | |
Collapse
|
21
|
Bayraktar S, Gutierrez-Barrera AM, Liu D, Tasbas T, Akar U, Litton JK, Lin E, Albarracin CT, Meric-Bernstam F, Gonzalez-Angulo AM, Hortobagyi GN, Arun BK. Outcome of triple-negative breast cancer in patients with or without deleterious BRCA mutations. Breast Cancer Res Treat 2011; 130:145-53. [PMID: 21830012 DOI: 10.1007/s10549-011-1711-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 07/27/2011] [Indexed: 12/31/2022]
Abstract
More than 75% of breast cancers that develop in BRCA1 mutation carriers are triple-negative breast cancers (TNBC). The aim of this study was to compare the recurrence-free survival (RFS) and overall survival (OS) in high-risk patients with TNBC with and without deleterious BRCA1/2 mutations. A total of 227 women with TNBC who were referred for genetic counseling and underwent BRCA genetic testing between 1997 and 2010 were included in the study. The relationships between clinical variables and outcomes were evaluated using univariate and multivariate Cox proportional hazard regression models. Of 227 high-risk women with TNBC, 50% (n = 114) tested positive for BRCA1/2 mutations. Age, race, and tumor characteristics did not differ between BRCA non-carriers and carriers. At a median follow-up of 3.4 years, the 5-year RFS rates were 74 and 81% (P = 0.21), and 5-year OS rates were 85 and 93% in BRCA non-carriers and BRCA carriers, respectively (P = 0.11). In a multivariate model, after adjusting for age and disease stage, BRCA carriers tended to have a decreased risk of recurrence (HR = 0.67; 95% CI: 0.38-1.19; P = 0.17) or death (HR = 0.51; 95% CI:0.23-1.17; P = 0.11) compared to non-carriers. Our data indicate a 50% prevalence of deleterious BRCA1/2 mutations in high-risk women diagnosed with TNBC. Overall prognosis of TNBC in BRCA carriers and non-carriers is not significantly different within the first 5 years following an initial diagnosis. Further studies need to evaluate whether different therapies will change the outcome in these subgroups of TNBC.
Collapse
Affiliation(s)
- Soley Bayraktar
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Alli E, Sharma VB, Hartman AR, Lin PS, McPherson L, Ford JM. Enhanced sensitivity to cisplatin and gemcitabine in Brca1-deficient murine mammary epithelial cells. BMC Pharmacol 2011; 11:7. [PMID: 21771338 PMCID: PMC3146825 DOI: 10.1186/1471-2210-11-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 07/19/2011] [Indexed: 12/01/2022] Open
Abstract
Background Breast cancers due to germline mutations or altered expression of the BRCA1 gene associate with an aggressive clinical course and frequently exhibit a "triple-negative" phenotype, i.e. lack of expression of the estrogen and progesterone hormone receptors and lack of overexpression of the HER2/NEU oncogene, thereby rendering them relatively insensitive to hormonal manipulation and targeted HER2 therapy, respectively. BRCA1 plays a role in multiple DNA repair pathways, and thus, when mutated, results in sensitivity to certain DNA damaging drugs. Results Here, we used a Brca1 murine mammary epithelial cell (MMEC) model to examine the effect of loss of Brca1 on cellular sensitivity to various chemotherapy drugs. To explore novel therapeutic strategies, we included DNA damaging and non-DNA damaging drugs whose mechanisms are dependent and independent of DNA repair, respectively, and drugs that are used in standard and non-standard lines of therapy for breast cancer. To understand the cellular mechanism, we also determined the role that DNA repair plays in sensitivity to these drugs. We found that cisplatin and gemcitabine had the greatest specific therapeutic benefit to Brca1-deficient MMECs, and that when used in combination produced a synergistic effect. This sensitivity may be attributed in part to defective NER, which is one of the DNA repair pathways normally responsible for repairing DNA adducts produced by cisplatin and is shown in this study to be defective in Brca1-deficient MMECs. Brca1-deficient MMECs were not differentially sensitive to the standard breast cancer chemotherapy drugs doxorubicin, docetaxel or 5-FU. Conclusions Both cisplatin and gemcitabine should be explored in clinical trials for first line regimens for BRCA1-associated and triple-negative breast cancer.
Collapse
Affiliation(s)
- Elizabeth Alli
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Center for Clinical Sciences Research, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|
23
|
Lee LJ, Alexander B, Schnitt SJ, Comander A, Gallagher B, Garber JE, Tung N. Clinical outcome of triple negative breast cancer in BRCA1 mutation carriers and noncarriers. Cancer 2011; 117:3093-100. [PMID: 21264845 PMCID: PMC4086795 DOI: 10.1002/cncr.25911] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2010] [Revised: 11/29/2010] [Accepted: 12/08/2010] [Indexed: 12/31/2022]
Abstract
BACKGROUND Women with BRCA1 mutations develop breast cancer with similar pathologic features to sporadic triple negative (TN) breast cancer, a subtype associated with early disease relapse and poor outcome. The clinical outcome of women with and without BRCA1 mutations who had TN breast cancer treated with conventional chemotherapy were compared. METHODS Women with stage I to III TN breast cancer who had BRCA1 testing within 36 months of diagnosis and received alkylating chemotherapy were identified from clinical databases and a Specialized Program of Research Excellence (SPORE) specimen bank. BRCA2 mutation carriers were excluded, resulting in a study cohort of 46 BRCA1 carriers and 71 noncarriers. Sites of metastasis, relapse rates, and survival were compared among carriers and noncarriers. The median follow-up was 75 months. RESULTS BRCA1 carriers were younger at diagnosis (P < .001) and had smaller tumors (P = .03) than noncarriers. Freedom from distant metastasis at 5 years was 76% for carriers and 70% for noncarriers (hazard ratio [HR] 0.79, P = .5). Sites of distant recurrence did not differ significantly (P = .15), although BRCA1 carriers had a propensity for brain relapse (58% vs 24%, P = .06). Overall survival at 5 years was 82% for carriers and 74% for noncarriers (HR 0.64, P = .25). Adjusting for age and stage, BRCA1 mutation status was not an independent predictor of survival (HR 0.73, P = .48). CONCLUSIONS BRCA1 mutation carriers with TN disease had similar survival rates to noncarriers when treated with alkylating chemotherapy. Women with BRCA1-related breast cancer may benefit from novel therapies that target DNA repair, and further study is needed to identify sporadic TN breast cancers with a BRCA-deficient phenotype.
Collapse
Affiliation(s)
- Larissa J. Lee
- Department of Radiation Oncology, Brigham and Women’s Hospital and Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Brian Alexander
- Department of Radiation Oncology, Brigham and Women’s Hospital and Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Stuart J. Schnitt
- Department of Pathology, Boston, MA
- Harvard Medical School, Boston, MA
| | - Amy Comander
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Bridget Gallagher
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA
| | - Judy E. Garber
- Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Nadine Tung
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
24
|
Hershko DD. Cyclin-dependent kinase inhibitor p27 as a prognostic biomarker and potential cancer therapeutic target. Future Oncol 2010; 6:1837-47. [PMID: 21142858 DOI: 10.2217/fon.10.144] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The prognosis and clinical management of patients with cancer is commonly determined by traditional clinical and pathological factors. Nevertheless, patients may present with significantly different clinical outcomes despite similar clinicopathological features. This has prompted intense research to find biological markers that may closely reflect tumor biology and thereby clinical outcome. This article presents the current knowledge on the prognostic significance of p27 expression in cancer and its potential role as a target for future therapy.
Collapse
Affiliation(s)
- Dan D Hershko
- Department of Surgery & Breast Health Institute, Rambam Health Care Campus & the Technion – Israel Institute of Technology, Haifa 31096, Israel
| |
Collapse
|
25
|
Norquist BM, Garcia RL, Allison KH, Jokinen CH, Kernochan LE, Pizzi CC, Barrow BJ, Goff BA, Swisher EM. The molecular pathogenesis of hereditary ovarian carcinoma: alterations in the tubal epithelium of women with BRCA1 and BRCA2 mutations. Cancer 2010; 116:5261-71. [PMID: 20665887 PMCID: PMC2975783 DOI: 10.1002/cncr.25439] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND BRCA1 or BRCA2 (BRCA1/2)-mutated ovarian carcinomas may originate in the fallopian tube. The authors of this report investigated alterations in BRCA1/2 tubal epithelium to define the molecular pathogenesis of these carcinomas. METHODS Tubal epithelium was evaluated from 31 BRCA1/2 mutation carriers with gynecologic carcinomas (BRCA CA), 89 mutation carriers who underwent risk-reducing salpingo-oophorectomy (RRSO), and 87 controls. Ki-67 expression and p53 foci (≥10 of 12 consecutive staining cells) were scored by 2 investigators who were blinded to patient designations. Expression levels of p27 and p21 were evaluated within p53 foci. Loss of heterozygosity at the BRCA1/2 mutation site was evaluated in microdissected p53 foci and tubal neoplasms. RESULTS Background tubal proliferation as measured by Ki-67 staining was increased in the BRCA1 RRSO group (P = .005) compared with the control group. Women who had BRCA1/2 mutations had more p53 foci identified per tubal segment than women in the control group (P = .02). Levels of p27 were decreased in 12 of 28 p53 foci from women with BRCA1 mutations and in 0 of 16 p53 foci from controls (P = .002). There was no loss of the wild type BRCA1/2 allele in 5 tested p53 foci. Tubal neoplasia lost the wild type allele in 6 of 6 foci (P = .002). CONCLUSIONS The current results suggested a model of tubal carcinogenesis in women with BRCA1/2 mutations. Increased proliferation occurred globally in at-risk tubal epithelium. A mutation in the tumor protein p53 gene TP53 with clonal proliferation and loss of p27 occurred before neoplastic proliferation. Loss of the wild type BRCA1/2 allele occurred with neoplastic proliferation and before invasion.
Collapse
Affiliation(s)
- Barbara M Norquist
- Department of Obstetrics and Gynecology, University of Washington School of Medicine, Seattle, WA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Coate L, Cuffe S, Horgan A, Hung RJ, Christiani D, Liu G. Germline genetic variation, cancer outcome, and pharmacogenetics. J Clin Oncol 2010; 28:4029-37. [PMID: 20679599 DOI: 10.1200/jco.2009.27.2336] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Studies of the role of germline or inherited genetic variation on cancer outcome can fall into three distinct categories. First, the impact of highly penetrant but lowly prevalent mutations of germline DNA on cancer prognosis has been studied extensively for BRCA1 and BRCA2 mutations as well as mutations related to hereditary nonpolyposis colorectal cancer syndrome. These mainly modest-sized analyses have produced conflicting results. Although some associations have been observed, they may not be independent of other known clinical or molecular prognostic factors. Second, the impact of germline polymorphisms on cancer prognosis is a burgeoning field of research. However, a deeper understanding of potentially confounding somatic changes and larger multi-institutional, multistage studies may be needed before consistent results are seen. Third, research examining the impact of germline genetic variation on differential treatment response or toxicity (pharmacogenetics) has produced some proof-of-principle results. Putative germline pharmacogenetic predictors of outcome include DPYD polymorphisms and fluorouracil toxicity, UGT1A1 variation and irinotecan toxicity, and CYP2D6 polymorphisms and tamoxifen efficacy, with emerging data on predictors of molecularly targeted or biologic drugs. Here we review data pertaining to these germline outcome and germline toxicity relationships.
Collapse
Affiliation(s)
- Linda Coate
- Department of Medical Oncology, Princess Margaret Hospital, 610 University Ave, Room 7-124, Toronto, Ontario, M5G 2M9 Canada
| | | | | | | | | | | |
Collapse
|
28
|
Taneja P, Maglic D, Kai F, Zhu S, Kendig RD, Fry EA, Inoue K. Classical and Novel Prognostic Markers for Breast Cancer and their Clinical Significance. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2010; 4:15-34. [PMID: 20567632 PMCID: PMC2883240 DOI: 10.4137/cmo.s4773] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The use of biomarkers ensures breast cancer patients receive optimal treatment. Established biomarkers such as estrogen receptor (ER) and progesterone receptor (PR) have been playing significant roles in the selection and management of patients for endocrine therapy. HER2 is a strong predictor of response to trastuzumab. Recently, the roles of ER as a negative and HER2 as a positive indicator for chemotherapy have been established. Ki67 has traditionally been recognized as a poor prognostic factor, but recent studies suggest that measurement of Ki67-positive cells during treatment will more effectively predict treatment efficacy for both anti-hormonal and chemotherapy. p53 mutations are found in 20–35% of human breast cancers and are associated with aggressive disease with poor clinical outcome when the DNA-binding domain is mutated. The utility of cyclin D1 as a predictor of breast cancer prognosis is controversial, but cyclin D1b overexpression is associated with poor prognosis. Likewise, overexpression of the low molecular weight form of cyclin E1 protein predicts poor prognosis. Breast cancers from BRCA1/2 carriers often show high nuclear grades, negativity to ER/PR/HER2, and p53 mutations, and thus, are associated with poor prognosis. The prognostic values of other molecular markers, such as p14ARF, TBX2/3, VEGF in breast cancer are also discussed. Careful evaluation of these biomarkers with current treatment modality is required to determine whether their measurement or monitoring offer significant clinical benefits.
Collapse
|
29
|
Guan X, Wang Y, Xie R, Chen L, Bai J, Lu J, Kuo MT. p27(Kip1) as a prognostic factor in breast cancer: a systematic review and meta-analysis. J Cell Mol Med 2010; 14:944-53. [PMID: 19298520 PMCID: PMC3823126 DOI: 10.1111/j.1582-4934.2009.00730.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The aim of this study was to comprehensively evaluate via a meta-analysis the association between p27 expression and clinical outcome in breast cancer patients. We conducted a meta-analysis of 20 studies (n= 6463 patients) that evaluated the correlation between p27 expression and indicators of breast cancer clinical outcome, including overall survival (OS), disease-free survival (DFS) and relapse-free survival (RFS). Data pooling was performed by RevMan 4.2. A total of 60% (9 of 15) of the studies showed a significant association between p27 high expression and OS, whereas 25% (2 of 8) and 60% (3 of 5) studies demonstrated a correlation between p27 high expression and DFS and RFS, respectively. The relative risks (RRs) were 1.34 (1.26–1.42) for OS (P < 0.00001), 1.27 (1.10–1.47) for DFS (P= 0.001) and 1.49 (0.92–2.42) for RFS (P= 0.10). In lymph node-negative breast cancer patients, the RRs for OS and RFS were 1.84 (1.30–2.59; P= 0.0005) and 1.30 (0.20–8.50; P= 0.78), respectively. In lymph node-positive breast cancer patients, the RRs for OS and RFS were 2.99 (1.77–5.07; P < 0.0001) and 1.49 (0.80–2.77; P= 0.21), respectively. This meta-analysis indicates that reduced p27 is an independent prognostic factor for poor overall and disease-free cancer survival.
Collapse
Affiliation(s)
- Xiaoxiang Guan
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing UniversityNanjing, Jiangsu, People’s Republic of China
- Department of Molecular Pathology, The University of Texas M. D. Anderson Cancer CenterHouston, TX, USA
- *Correspondence to: Longbang CHEN, M.D., Ph.D, Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing 210002, People’s Republic of China. Tel.: +86-25-80860123 Fax: +86-25-84824051 E-mail:
| | - Yucai Wang
- Department of Experimental Radiation Oncology, The University of Texas M. D. Anderson Cancer CenterHouston, TX, USA
- The University of Texas Graduate School of Biomedical Sciences at HoustonHouston, TX, USA
| | - Ruilian Xie
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing UniversityNanjing, Jiangsu, People’s Republic of China
| | - Longbang Chen
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing UniversityNanjing, Jiangsu, People’s Republic of China
- *Correspondence to: Longbang CHEN, M.D., Ph.D, Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing 210002, People’s Republic of China. Tel.: +86-25-80860123 Fax: +86-25-84824051 E-mail:
| | - Jianling Bai
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical UniversityNanjing, Jiangsu, People’s Republic of China
| | - Jia Lu
- Department of Molecular Pathology, The University of Texas M. D. Anderson Cancer CenterHouston, TX, USA
| | - Macus Tien Kuo
- Department of Molecular Pathology, The University of Texas M. D. Anderson Cancer CenterHouston, TX, USA
| |
Collapse
|
30
|
Prognosis of BRCA-associated breast cancer: a summary of evidence. Breast Cancer Res Treat 2009; 119:13-24. [DOI: 10.1007/s10549-009-0566-z] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Accepted: 09/17/2009] [Indexed: 01/04/2023]
|
31
|
Filipits M, Rudas M, Heinzl H, Jakesz R, Kubista E, Lax S, Schippinger W, Dietze O, Greil R, Stiglbauer W, Kwasny W, Nader A, Stierer M, Gnant MFX. Low p27 expression predicts early relapse and death in postmenopausal hormone receptor-positive breast cancer patients receiving adjuvant tamoxifen therapy. Clin Cancer Res 2009; 15:5888-94. [PMID: 19723645 DOI: 10.1158/1078-0432.ccr-09-0728] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Previously, we have shown that p27 may be a potential predictive biomarker for the selection of premenopausal women with early-stage hormone-responsive breast cancer for adjuvant endocrine therapy. The purpose of the present study was to assess the clinical relevance of p27 expression in postmenopausal hormone receptor-positive breast cancer patients who were treated with adjuvant tamoxifen therapy. EXPERIMENTAL DESIGN We determined the expression of p27 by immunohistochemistry in the surgical specimens of breast carcinoma patients who had been enrolled in Austrian Breast and Colorectal Cancer Study Group Trial 06 and received tamoxifen for 5 years. Early relapse and death within the first 5 years of follow-up were analyzed using Cox models adjusted for clinical and pathologic factors. RESULTS p27 expression was high (>70% p27-positive tumor cells) in 252 of 483 (52%) tumor specimens and was associated with favorable outcome of the patients. Women with high p27 expression had a significantly longer disease-free survival (adjusted hazard ratio for relapse, 0.22; 95% confidence interval, 0.11-0.42; P < 0.001) and overall survival (adjusted hazard ratio for death, 0.39; 95% confidence interval, 0.21-0.72; P = 0.002) as compared with women with low p27 expression. CONCLUSION Low p27 expression independently predicts early relapse and death in postmenopausal women with early-stage, hormone receptor-positive breast cancer who received adjuvant tamoxifen for 5 years.
Collapse
Affiliation(s)
- Martin Filipits
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
van der Groep P, van Diest PJ, Menko FH, Bart J, de Vries EGE, van der Wall E. Molecular profile of ductal carcinoma in situ of the breast in BRCA1 and BRCA2 germline mutation carriers. J Clin Pathol 2009; 62:926-30. [DOI: 10.1136/jcp.2009.065524] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Aims:Ductal carcinoma in situ (DCIS) is an established late precursor of sporadic invasive breast cancer and to a large extent parallels its invasive counterpart with respect to molecular changes and immunophenotype. Invasive breast cancers in germline BRCA1 and BRCA2 mutation carriers have a distinct “basal” and “luminal” immunophenotype, respectively, but the immunophenotype of their precursor lesions has hardly been studied, and this was the aim of this study.Methods:DCIS lesions of 25 proven BRCA1 and 9 proven BRCA2 germline mutation carriers and their 22 and 6, respectively, accompanying invasive lesions were stained by immunohistochemistry for oestrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor (HER)2/neu, cytokeratin (CK)5/6, CK14, epidermal growth factor receptor (EGFR) and Ki67.Results:DCIS lesions in BRCA1 mutation carriers were mostly of the basal molecular type with low ER/PR/HER2 expression, while they frequently expressed CK5/6, CK14 and EGFR, and were mostly grade 3 and highly proliferative. DCIS lesions in BRCA2 mutation carriers were mostly of luminal molecular type with frequent expression of ER/PR, and infrequent expression of CK5/6, CK14 and EGFR, and they were mostly grade 3 and showed low proliferation. In BRCA1 and BRCA2 mutation carriers there was a high concordance between DCIS lesions and their concomitant invasive counterpart with regard to expression of individual markers as well as “molecular” subtype.Conclusions:Although the number of cases studied was low, DCIS lesions in BRCA1 and BRCA2 mutations carriers are usually of the basal and luminal molecular type, respectively, similar to their accompanying invasive cancers, thereby providing evidence that DCIS is a direct precursor lesion in these hereditary predisposed patients. This also suggests that crucial carcinogenetic events leading to these phenotypes in hereditary predisposed patients occur before the stage of invasion.
Collapse
|
33
|
Dworkin AM, Huang THM, Toland AE. Epigenetic alterations in the breast: Implications for breast cancer detection, prognosis and treatment. Semin Cancer Biol 2009; 19:165-71. [PMID: 19429480 PMCID: PMC2734184 DOI: 10.1016/j.semcancer.2009.02.007] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Accepted: 02/11/2009] [Indexed: 12/24/2022]
Abstract
Epigenetic alterations of the genome such as DNA promoter methylation and chromatin remodeling play an important role in tumorigenesis. Recent findings indicate epigenetic modifications as key factors in breast carcinogenesis. These modifications are quite appealing as targets for preventative care and therapeutics because of their potential for reversal. Future medical care for breast cancer patients will likely depend upon a better understanding of the roles epigenetic modifications play in carcinogenesis. Here, we discuss the importance of epigenetics in breast cancer detection, prognosis, and therapy with an emphasis on mechanisms and epigenetic contributions to field cancerization effects.
Collapse
Affiliation(s)
- Amy M Dworkin
- Integrated Biomedical Science Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | | | | |
Collapse
|
34
|
Xu X, Gammon MD, Zhang Y, Bestor TH, Zeisel SH, Wetmur JG, Wallenstein S, Bradshaw PT, Garbowski G, Teitelbaum SL, Neugut AI, Santella RM, Chen J. BRCA1 promoter methylation is associated with increased mortality among women with breast cancer. Breast Cancer Res Treat 2009; 115:397-404. [PMID: 18521744 PMCID: PMC2693263 DOI: 10.1007/s10549-008-0075-5] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Accepted: 05/19/2008] [Indexed: 12/20/2022]
Abstract
Promoter-CpG island hypermethylation has been proposed as an alternative mechanism to inactivate BRCA1 in the breast where somatic mutations of BRCA1 are rare. To better understand breast cancer etiology and progression, we explored the association between BRCA1 promoter methylation status and prognostic factors as well as survival among women with breast cancer. Promoter methylation of BRCA1 was assessed in 851 archived tumor tissues collected from a population-based study of women diagnosed with invasive or in situ breast cancer in 1996-1997, and who were followed for vital status through the end of 2002. About 59% of the tumors were methylated at the promoter of BRCA1. The BRCA1 promoter methylation was more frequent in invasive cancers (P = 0.02) and among premenopausal cases (P = 0.05). BRCA1 promoter methylation was associated with increased risk of breast cancer-specific mortality (age-adjusted HR 1.71; 95% CI: 1.05-2.78) and all-cause mortality (age-adjusted HR 1.49; 95% CI: 1.02-2.18). Neither dietary methyl intakes in the year prior to the baseline interview nor the functional polymorphisms in one-carbon metabolism were associated with BRCA1 methylation status. Our study is the first epidemiological investigation on the prognostic value of BRCA1 promoter methylation in a large population-based cohort of breast cancer patients. Our results indicate that BRCA1 promoter methylation is an important factor to consider in predicting breast cancer survival.
Collapse
Affiliation(s)
- Xinran Xu
- Department of Community and Preventive Medicine, Mount Sinai School of Medicine, New York, NY 10029
| | - Marilie D. Gammon
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599
| | - Yujing Zhang
- Department of Environmental Health Sciences, Columbia University, New York, NY 10032
| | - Timothy H. Bestor
- Department of Genetics and Development, Columbia University, New York, NY 10032
| | - Steven H. Zeisel
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27599
| | - James G. Wetmur
- Department of Microbiology, Mount Sinai School of Medicine, New York, NY 10029
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY 10029
| | - Sylvan Wallenstein
- Department of Community and Preventive Medicine, Mount Sinai School of Medicine, New York, NY 10029
| | - Patrick T. Bradshaw
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599
| | - Gail Garbowski
- Department of Environmental Health Sciences, Columbia University, New York, NY 10032
| | - Susan L. Teitelbaum
- Department of Community and Preventive Medicine, Mount Sinai School of Medicine, New York, NY 10029
| | - Alfred I. Neugut
- Department of Epidemiology, Columbia University, New York, NY 10032
- Department of Medicine, Columbia University, New York, NY 10032
| | - Regina M. Santella
- Department of Environmental Health Sciences, Columbia University, New York, NY 10032
| | - Jia Chen
- Department of Community and Preventive Medicine, Mount Sinai School of Medicine, New York, NY 10029
- Department of Pediatrics, Mount Sinai School of Medicine, New York, NY 10029
- Department of Oncological Science, Mount Sinai School of Medicine, New York, NY 10029
| |
Collapse
|
35
|
Spurdle AB, Deans AJ, Duffy D, Goldgar DE, Chen X, Beesley J, kConFaB, Easton DF, Antoniou AC, Peock S, Cook M, EMBRACE Study Collaborators, Nathanson KL, Domchek SM, MacArthur GA, Chenevix-Trench G. No evidence that CDKN1B (p27) polymorphisms modify breast cancer risk in BRCA1 and BRCA2 mutation carriers. Breast Cancer Res Treat 2009; 115:307-13. [PMID: 18543099 DOI: 10.1007/s10549-008-0083-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Accepted: 05/27/2008] [Indexed: 11/25/2022]
Abstract
The p27(kip1) protein functions as an inhibitor of cyclin dependent kinase-2, and shows loss of expression in a large percentage of BRCA1 and BRCA2 breast cancer cases. We investigated the association between CDKN1B gene variants and breast cancer risk in 2359 female BRCA1 and BRCA2 mutation carriers from Australia, the UK, and the USA. Samples were genotyped for five single nucleotide polymorphisms, including coding variant rs2066827 (V109G). Cox regression provided no convincing evidence that any of the polymorphisms modified disease risk for BRCA1 or BRCA2 carriers, either alone or as a haplotype. Borderline associations were observed for homozygote carriers of the rs3759216 rare allele, but were opposite in effect for BRCA1 and BRCA2 carriers (adjusted hazard ratio (HR) 0.72 (95% CI = 0.53-0.99; P = 0.04 for BRCA1, HR 1.47 (95% CI = 0.99-2.18; P = 0.06 for BRCA2). The 95% confidence intervals for per allele risk estimates excluded a twofold risk, indicating that common CDKN1B polymorphisms do not markedly modify breast cancer risk among BRCA1 or BRCA2 carriers.
Collapse
Collaborators
Susan Peock, Margaret Cook, Alexandra Bignell, Debra Frost, Neva Haites, Helen Gregory, Patrick Morrison, Trevor Cole, Carole McKeown, Alan Donaldson, Joan Paterson, Alexandra Murray, Mark Rogers, Emma McCann, Peter Daly, David Barton, Mary Porteous, Michael Steel, Carole Brewer, Julia Rankin, Rosemarie Davidson, Victoria Murday, Louise Izatt, Gabriella Pichert, Huw Dorkins, Richard Trembath, Tim Bishop, Carol Chu, Ian Ellis, D Gareth Evans, Fiona Lalloo, Alison Male, James Mackay, Anne Robinson, Carol Gardiner, Fiona Douglas, John Burn, Lucy Side, Lisa Walker, Sarah Durell, Rosalind Eeles, Jackie Cook, Oliver Quarrell, Shirley Hodgson, Diana Eccles, Anneke Lucassen,
Collapse
|
36
|
Hagen AI, Tretli S, Maehle L, Apold J, Vedå N, Møller P. Survival in Norwegian BRCA1 mutation carriers with breast cancer. Hered Cancer Clin Pract 2009; 7:7. [PMID: 19366445 PMCID: PMC2678098 DOI: 10.1186/1897-4287-7-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Accepted: 04/14/2009] [Indexed: 11/24/2022] Open
Abstract
Several studies of survival in women with BRCA1 mutations have shown either reduced survival or no difference compared to controls. Programmes for early detection and treatment of inherited breast cancer, have failed to demonstrate a significant improvement in survival in BRCA1 mutation carriers. One hundred and sixty-seven women with disease-associated germline BRCA1 mutations and breast cancer from 1980 to 2001 were identified. Tumour characteristics, treatment given and survival were recorded. A control group comprising three hundred and four women matched for age, time of diagnosis and stage were used to compare survival. BRCA1 mutation carriers were found to have a poorer prognosis, which could be explained by neither the mode of surgical treatment nor the use of adjuvant chemotherapy. BRCA1 mutation carriers with node negative breast cancer had worse overall survival than controls. Our findings confirm the serious prognosis of BRCA1-associated breast cancer even when diagnosed at an early stage, and that type of treatment does not influence prognosis.
Collapse
Affiliation(s)
- Anne Irene Hagen
- Institute of clinical medicine, University of Bergen, N-5021 Bergen, Norway.
| | | | | | | | | | | |
Collapse
|
37
|
Rakha EA, Elsheikh SE, Aleskandarany MA, Habashi HO, Green AR, Powe DG, El-Sayed ME, Benhasouna A, Brunet JS, Akslen LA, Evans AJ, Blamey R, Reis-Filho JS, Foulkes WD, Ellis IO. Triple-negative breast cancer: distinguishing between basal and nonbasal subtypes. Clin Cancer Res 2009; 15:2302-10. [PMID: 19318481 DOI: 10.1158/1078-0432.ccr-08-2132] [Citation(s) in RCA: 364] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE Triple-negative (TN; estrogen receptor, progesterone receptor, and HER-2 negative) cancer and basal-like breast cancer (BLBC) are associated with poor outcome and lack the benefit of targeted therapy. It is widely perceived that BLBC and TN tumors are synonymous and BLBC can be defined using a TN definition without the need for the expression of basal markers. EXPERIMENTAL DESIGN We have used two well-defined cohorts of breast cancers with a large panel of biomarkers, BRCA1 mutation status, and follow-up data to compare the clinicopathologic and immunohistochemical features of TN tumors expressing one or more of the specific basal markers (CK5/6, CK17, CK14, and epidermal growth factor receptor; BLBC) with those TN tumors that express none of these markers (TN3BKE-). RESULTS Here, we show that although the morphologic features of BLBC are not significantly different from that of TN3BKE- tumors, BLBC showed distinct clinical and immunophenotypic differences. BLBC showed a statistically significant association with the expression of the hypoxia-associated factor (CA9), neuroendocrine markers, and other markers of poor prognosis such as p53. A difference in the expression of cell cycle-associated proteins and biomarkers involved in the immunologic portrait of tumors was seen. Compared with TN3BKE- tumors, BLBC was positively associated with BRCA1 mutation status and showed a unique pattern of distant metastasis, better response to chemotherapy, and shorter survival. CONCLUSION TN breast cancers encompass a remarkably heterogeneous group of tumors. Expression of basal markers identifies a biologically and clinically distinct subgroup of TN tumors, justifying the use of basal markers (in TN tumors) to define BLBC.
Collapse
Affiliation(s)
- Emad A Rakha
- Department of Histopathology, Nottingham City Hospital NHS Trust, Nottingham University, Nottingham, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Voduc D, Nielsen TO, Cheang MC, Foulkes WD. The combination of high cyclin E and Skp2 expression in breast cancer is associated with a poor prognosis and the basal phenotype. Hum Pathol 2008; 39:1431-7. [DOI: 10.1016/j.humpath.2008.03.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2007] [Revised: 03/05/2008] [Accepted: 03/11/2008] [Indexed: 10/21/2022]
|
39
|
Oseni T, Jatoi I. An Overview of the Role of Prophylactic Surgery in the Management of Individuals with a Hereditary Cancer Predisposition. Surg Clin North Am 2008; 88:739-58, vi. [DOI: 10.1016/j.suc.2008.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
40
|
Chu IM, Hengst L, Slingerland JM. The Cdk inhibitor p27 in human cancer: prognostic potential and relevance to anticancer therapy. Nat Rev Cancer 2008; 8:253-67. [PMID: 18354415 DOI: 10.1038/nrc2347] [Citation(s) in RCA: 774] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The cyclin-dependent kinase (Cdk) inhibitor p27 (also known as KIP1) regulates cell proliferation, cell motility and apoptosis. Interestingly, the protein can exert both positive and negative functions on these processes. Diverse post-translational modifications determine the physiological role of p27. Phosphorylation regulates p27 binding to and inhibition of cyclin-Cdk complexes, its localization and its ubiquitin-mediated proteolysis. In cancers, p27 is inactivated through impaired synthesis, accelerated degradation and by mislocalization. Moreover, studies in several tumour types indicate that p27 expression levels have both prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Isabel M Chu
- Braman Family Breast Cancer Institute, and Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, Miami, Florida 33136, USA
| | | | | |
Collapse
|
41
|
Arnes JB, Collett K, Akslen LA. Independent prognostic value of the basal-like phenotype of breast cancer and associations with EGFR and candidate stem cell marker BMI-1. Histopathology 2008; 52:370-80. [PMID: 18269588 DOI: 10.1111/j.1365-2559.2007.02957.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIMS To study the relationship between basal-like breast cancers, epidermal growth factor receptor (EGFR) and candidate stem cell markers (BMI-1, EZH2, Oct-4) in a population-based setting. METHODS AND RESULTS Immunohistochemistry was evaluated in a series of 190 breast cancers. Basal-like phenotype (BLP) 1-5 was found in 4.3-14.3% of cases. EGFR was expressed in 9% of cases and associated with cytokeratin (CK) 5 and P-cadherin positivity, but not with survival; 28% of CK5+ cases were EGFR+. On multivariate analysis, basal-like differentiation and lymph node status were independent prognostic factors of comparable strength. BMI-1 positivity (42.6%) was associated with absence of basal-like features, oestrogen receptor positivity and low Ki67, but not related to survival. BMI was not associated with EZH2 expression, and these markers tended to show opposite associations with other variables, suggesting different roles in breast cancer. Oct-4 expression was not detected in this series. CONCLUSIONS Basal-like features and lymph node status were strong and independent prognostic factors in this population-based series of breast cancer. Neither EGFR nor BMI-1 had significant prognostic impact, whereas EZH2 expression was associated with decreased survival. BMI-1 was inversely related to basal-like factors, and a stem cell phenotype of the basal-like subgroup could not be verified by this marker.
Collapse
Affiliation(s)
- J B Arnes
- The Gade Institute, Section for Pathology, University of Bergen, Haukeland University Hospital, Bergen, Norway
| | | | | |
Collapse
|
42
|
Biomarkers for Breast Cancer: Towards the Proposition of Clinically Relevant Tools. Breast Cancer 2007. [DOI: 10.1007/978-3-540-36781-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
43
|
Honrado E, Osorio A, Milne RL, Paz MF, Melchor L, Cascón A, Urioste M, Cazorla A, Díez O, Lerma E, Esteller M, Palacios J, Benítez J. Immunohistochemical classification of non-BRCA1/2 tumors identifies different groups that demonstrate the heterogeneity of BRCAX families. Mod Pathol 2007; 20:1298-306. [PMID: 17885670 DOI: 10.1038/modpathol.3800969] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Around 25% of hereditary breast and ovarian cancer families have mutations in the BRCA1 and BRCA2 genes. The search for other genes has until now failed, probably because there is not one single BRCAX gene, but rather various genes that may each be responsible for a small number of breast cancer families and/or may interact according to a polygenic model. We have studied 50 tumors from probands belonging to non-BRCA1/2 breast cancer families (BRCAX), using 25 immunohistochemical markers. The objective was to classify these tumors and confirm that they are heterogeneous. Unsupervised cluster analysis showed the existence of the following two main groups of tumors: high-grade and estrogen receptor (ER)-negative tumors (50%), and low-grade and ER-positive tumors (50%). In addition we identified five subgroups, three among the high-grade and two among the low-grade groups; one overexpressing HER-2 (18%); one with a basal-like phenotype (14%); one with a normal breast-like phenotype (18%); a luminal A subgroup (36%), and a luminal B subgroup (14%). Hypermethylation of the BRCA1 gene was observed in 42% of the cases, spread across all five subgroups, but only 37% of those had loss of heterozygosity as well. These latter cases were all clustered in the high-grade group and the majority of them in the basal-like subgroup. Our results show that familial non-BRCA1/2 tumors are heterogeneous and suggest a polygenic model for explaining the majority of BRCAX families. In addition we have defined a subset of them that have somatic inactivation of the BRCA1 gene.
Collapse
Affiliation(s)
- Emiliano Honrado
- Department of Human Genetics, Spanish National Cancer Centre, Melchor Fernandez Almagro 3, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
High frequency of HIF-1alpha overexpression in BRCA1 related breast cancer. Breast Cancer Res Treat 2007; 111:475-80. [PMID: 18030615 DOI: 10.1007/s10549-007-9817-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2007] [Accepted: 11/08/2007] [Indexed: 10/22/2022]
Abstract
Hypoxia is a hallmark of cancer. Hypoxia inducible factor-1alpha (HIF-1alpha) is the key regulator of the hypoxia response. HIF-1alpha is overexpressed during sporadic breast carcinogenesis and correlated with poor prognosis. Little is known on the role of HIF-1alpha in hereditary breast carcinogenesis. A recent study suggests a role for BRCA1 in HIF-1alpha regulation. We therefore examined the expression of HIF-1alpha in BRCA1 related breast cancers. By immunohistochemistry we studied expression of HIF-1alpha and some of its downstream targets in 30 hereditary invasive breast cancers in comparison with 200 sporadic controls. HIF-1alpha overexpression was significantly more frequent in BRCA1 related breast cancers (27/30, 90%) than in sporadic controls (88/200, 44%) (P < 0.0001). 19/30 (63%) of BRCA1 tumors showed perinecrotic (hypoxia induced) and 8/30 (27%) a diffuse HIF-1alpha overexpression pattern, the latter more likely related to genetic alterations in oncogenes and tumor suppressor genes. In contrast, sporadic breast cancer HIF-1 expressing tumors showed an inverse ratio of perinecrotic/diffuse expression (31 vs. 69%, P = 0.0002). Glut-1 and CAIX, downstream HIF1 targets, were expressed in 27/30 (90%) and 15/21 (71%) of hereditary cases versus 54/183 (29%) and 24/183 (13%) in sporadic cases. p300 levels, necessary for HIF-1 downstream activation, were significantly higher in hereditary cases (20/21, 95%) compared to sporadic cases (91/183, 50%, P = 0.0001). In conclusion, in BRCA1 germline mutation related breast cancer, functional HIF-1alpha overexpression is seen at a much higher frequency than in sporadic breast cancer, mostly hypoxia induced. This points to an important role of hypoxia and its key regulator HIF-1alpha in hereditary breast carcinogenesis.
Collapse
|
45
|
Abstract
Mutations in BRCA1 and BRCA2 are well-established causes of hereditary breast cancer. As genetic testing becomes more widespread, increasing numbers of women are known to have mutations at or shortly after their breast cancer diagnosis. Current evidence is insufficient to mandate different local or systemic treatment based upon the presence of a germline mutation. The well-documented increased risk of contralateral second primary breast cancer and possibly of late ipsilateral second primary breast cancers may influence patient decision-making with regard to breast-conserving treatment.
Collapse
Affiliation(s)
- Mark E Robson
- Clinical Genetics and Breast Cancer Medicine Services, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA.
| |
Collapse
|
46
|
Rubinstein WS. Hereditary breast cancer: pathobiology, clinical translation, and potential for targeted cancer therapeutics. Fam Cancer 2007; 7:83-9. [PMID: 17624601 DOI: 10.1007/s10689-007-9147-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2007] [Accepted: 05/14/2007] [Indexed: 11/24/2022]
Abstract
BRCA1 and BRCA2 breast cancers have distinct biological features as evidenced by histopathologic, immunohistochemical, gene expression profiling, and array-comparative genomic hybridization data. BRCA1 breast cancers may have a worse prognosis but may, however be amenable to treatment such as chemotherapy for small high-grade, lymph node negative breast cancers. Paradoxically, tamoxifen may provide effective adjuvant and chemopreventive therapy despite the predominantly negative estrogen receptor status of BRCA1 breast cancers. The distinctive biology of BRCA1 and BRCA2 breast cancers bodes well for the development of targeted cancer therapies. Cells with BRCA1 or BRCA2 loss of function are deficient in DNA double strand break repair and are sensitized to poly(ADP-ribose) polymerase (PARP) inhibitors, causing the persistence of DNA lesions which are usually repaired by homologous recombination and ultimately leading to apoptosis. The potentially high efficacy and low toxicity of poly(ADP-ribose) polymerase inhibitors presents an opportunity for targeted cancer therapeutics for BRCA1 and BRCA2 germline mutation carriers. Genotype-tailored chemoprevention may be feasible which could theoretically eliminate single cells that have sustained a second hit, before cancer progression takes place. If targeted cancer therapies emerge, it will become crucially important to identify BRCA carriers at the time of diagnosis for optimal therapy and to identify unaffected carriers for chemoprevention. If so, then to the extent that barriers in the recognition and referral of patients to genetic counseling cannot be surmounted, pathological and genomic methods to identify a BRCA1 or BRCA2 breast cancer profile will gain increasing clinical importance.
Collapse
|
47
|
Su Y, Vanderlaag K, Ireland C, Ortiz J, Grage H, Safe S, Frankel AE. 1,1-Bis(3'-indolyl)-1-(p-biphenyl)methane inhibits basal-like breast cancer growth in athymic nude mice. Breast Cancer Res 2007; 9:R56. [PMID: 17764562 PMCID: PMC2206732 DOI: 10.1186/bcr1761] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2007] [Revised: 07/18/2007] [Accepted: 08/31/2007] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION 1,1-Bis (3'-indolyl)-1-(p-biphenyl) methane (CDIM9) has been identified as a new peroxisome proliferator-activated receptor (PPAR)-gamma agonist that exhibits both receptor dependent and independent antitumor activities. CDIM9 has not previously been studied with respect to its effects against basal-like breast cancer. Our goal in the present study was to investigate the anti-basal-like breast tumor activity of CDIM9 in vitro and in vivo. METHODS The effects of CDIM9 on cell protein and DNA syntheses were determined in basal-like breast cancer MDA-MB231 and BT549 cells in vitro. Maximum tolerated dose and dose-limited toxicity were determined in BalB/c mice, and antitumor growth activities were assessed in MDA-MB231 basal-like breast tumor xenografts in athymic nude mice. RESULTS CDIM9 exhibited selective cell cytotoxicity and anti-proliferation effects on basal-like breast cancer lines. In MDA-MB231 cell, CDIM9 induced caveolin-1 and p27 expression, which was significantly downregulated by co-treatment with the PPAR-gamma antagonist GW9662. Nonsteroidal anti-inflammatory drug-activated gene-1 and activating transcription factor-3 were upregulated by CDIM9 through a PPAR-gamma independent pathway. CDIM9 (40 mg/kg daily, intraperitoneally, for 35 days) inhibited the growth of subcutaneous MDA-MB231 tumor xenografts by 87%, and produced a corresponding decrease in proliferation index. Nearly half of the treated mice (46%) had complete durable remissions, confirmed by histology. The growth of an established tumor was inhibited by CDIM9 treatment (64 mg/kg daily, intraperitoneally, for 10 days), with a mean tumor growth inhibition of 67% as compared with controls. CDIM9 induced increases in tumor caveolin-1 and p27 in vivo, which may contribute to its antitumor activity in basal-like breast cancer. CONCLUSION CDIM9 showed potent antiproliferative effects on basal-like breast cancer cell in tissue culture and dramatic growth inhibition in animal models at safe doses. These findings justify further development of this drug for treatment of basal-like breast cancer.
Collapse
Affiliation(s)
- Yunpeng Su
- Scott & White Cancer Research Institute, South Airport Road, Temple, Texas 76502, USA
| | - Kathryn Vanderlaag
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466
| | - Courtney Ireland
- Scott & White Cancer Research Institute, South Airport Road, Temple, Texas 76502, USA
| | - Janelle Ortiz
- Scott & White Cancer Research Institute, South Airport Road, Temple, Texas 76502, USA
| | - Henry Grage
- Plantacor, Inc., 526 University Dr. East Suite 101A, College Station, Texas 77840 USA
| | - Stephen Safe
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466
| | - Arthur E Frankel
- Scott & White Cancer Research Institute, South Airport Road, Temple, Texas 76502, USA
| |
Collapse
|
48
|
Liebens FP, Carly B, Pastijn A, Rozenberg S. Management of BRCA1/2 associated breast cancer: a systematic qualitative review of the state of knowledge in 2006. Eur J Cancer 2006; 43:238-57. [PMID: 17095205 DOI: 10.1016/j.ejca.2006.07.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2006] [Revised: 07/02/2006] [Accepted: 07/28/2006] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The optimal clinical management of breast cancer (BC) arising in BRCA1/2 mutations carriers is a difficult issue complicated by the risk of subsequent malignancies and by the potential differences in response to local and systemic therapies. AIM Systematically review the difference in outcome after breast conservation therapy (BCT) and uni-or bilateral mastectomy in BRCA1/2 related BC. MATERIAL AND METHODS We selected 20 studies, for which we evaluated the methodology, the characteristics of the populations, biases, confounding risk factors and outcomes. RESULTS All studies are retrospective, entailed by numerous biases. They varied with respect to patients' number, selection, and confounding factors. Hereditary BC patients carried an increased risk of ipsilateral recurrence in 5/17 studies, a worse survival in 4/14, an increased risk of contralateral BC in 14/16. CONCLUSION Except for contralateral risk, the presence of a BRCA mutation does not seem to offer additional prognostic information. Large prospective trials, stratified for risk reduction strategies are warranted.
Collapse
|
49
|
Elzagheid A, Kuopio T, Pyrhönen S, Collan Y. Lymph node status as a guide to selection of available prognostic markers in breast cancer: the clinical practice of the future? Diagn Pathol 2006; 1:41. [PMID: 17092354 PMCID: PMC1654187 DOI: 10.1186/1746-1596-1-41] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2006] [Accepted: 11/08/2006] [Indexed: 11/10/2022] Open
Abstract
Prognosticators evaluating survival in breast cancer vary in significance in respect to lymph node status. Studies have shown e.g. that HER2/neu immunohistochemistry or HER2/neu gene amplification analysis do perform well as prognosticators in lymph node positive (LN +) patients but are less valuable in lymph node negative (LN -) patients. We collected data from different studies and tried to evaluate the relative significance of different prognosticators in LN+/LN- patient groups. In LN+ patients HER2/neu and E-cadherin immunohistochemistry were the statistically most significant prognosticators followed by proliferation associated features (mitotic counts by SMI (standardised mitotic index) or MAI (mitotic activity index), or S-phase fraction). Bcl-2 immunohistochemistry was also significant but p53 and cystatin A had no significance as prognosticators. In LN- patients proliferation associated prognosticators (SMI, MAI, Ki-67 index, PCNA immunohistochemistry, S-phase fraction) are especially valuable and also Cathepsin D, cystatin A, and p53 are significant, but HER2/neu or bcl-2, or E-cadherin less significant or without significance. We find that in studies evaluating single prognosticators one should distinguish between prognosticators suitable for LN+ and LN- patients. This will allow the choice of best prognosticators in evaluating the prospects of the patient. The distinction between LN+ and LN- patients in this respect may also be of special value in therapeutic decisions.
Collapse
Affiliation(s)
- A Elzagheid
- Department of Oncology and Radiotherapy, Turku University Hospital, Savitehtaankatu 1 PB 52, FIN-20521, Turku, Finland
- Department of Pathology, University of Turku, and Turku University Hospital, Kiinamyllynkatu 10, FIN-20520, Turku, Finland
| | - T Kuopio
- Department of Pathology, Jyväskylä Central Hospital, FIN-40620, Jyväskylä, Finland
| | - S Pyrhönen
- Department of Oncology and Radiotherapy, Turku University Hospital, Savitehtaankatu 1 PB 52, FIN-20521, Turku, Finland
| | - Y Collan
- Department of Pathology, University of Turku, and Turku University Hospital, Kiinamyllynkatu 10, FIN-20520, Turku, Finland
| |
Collapse
|
50
|
Bonadona V, Dussart-Moser S, Voirin N, Sinilnikova OM, Mignotte H, Mathevet P, Brémond A, Treilleux I, Martin A, Romestaing P, Raudrant D, Rudigoz RC, Lenoir GM, Lasset C. Prognosis of early-onset breast cancer based on BRCA1/2 mutation status in a French population-based cohort and review. Breast Cancer Res Treat 2006; 101:233-45. [PMID: 17061047 DOI: 10.1007/s10549-006-9288-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2006] [Accepted: 05/24/2006] [Indexed: 10/24/2022]
Abstract
PURPOSE The debate concerning poorer survival for patients with breast cancer (BC) carrying a BRCA1 germline mutation is unresolved, and requires additional data from population-based studies. PATIENTS AND METHODS We followed 232 women with invasive BC under age 46, ascertained prospectively through a French population-based BC registry and tested for BRCA1/2 mutations (median follow-up: 82 months). We compared tumour characteristics and survival rates between 21 BRCA1/2 deleterious mutation carriers and 211 non-carriers. RESULTS As compared to sporadic tumours, BRCA1/2 tumours showed higher grade (P = 0.02), fewer ductal carcinoma in situ (P = 0.02), more frequent medullary histology (P = 0.02), more frequent negative oestrogen and progesterone receptors (P = 0.001 each). At 5 years, BC-specific survival, metastasis-free survival, ipsilateral recurrence-free survival and contralateral BC-free survival rates for BRCA1/2 mutation carriers were 95.0%, 94.7%, 100% and 90.0% respectively, compared with 89.6%, 78.2%, 88.8% and 94.4% respectively, for non-carriers (not significant). Rates for women carrying only a BRCA1 mutation were 93.3%, 93.3%, 100%, 86.7%, respectively. 76% of BRCA1/2 carriers received chemotherapy. CONCLUSION Despite unfavourable tumour features, we found no evidence for poorer short-term survival in BRCA1 mutation carriers compared to non-carriers in this prospective population-based cohort. The high rate of BRCA1 carriers who received chemotherapy for their BC should question the positive impact of this treatment, as suggested by preclinical studies showing increased chemosensitivity of BRCA1-associated tumours.
Collapse
Affiliation(s)
- Valérie Bonadona
- Centre Léon Bérard, Unit of Prevention and Genetic Epidemiology, 28 rue Laënnec, 69 373 Lyon cedex 08, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|