1
|
Green MD, Brenneman R, Powell SN, Bergom C. Harnessing the DNA Repair Pathway in Breast Cancer: Germline Mutations/Polymorphisms in Breast Radiation. Semin Radiat Oncol 2022; 32:298-302. [PMID: 35688528 DOI: 10.1016/j.semradonc.2022.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Molecular profiling facilitates opportunities for personalization of breast cancer management. Increasing availability of germline and somatic sequencing provides insight into predictors of treatment efficacy and treatment tolerance of patients with breast cancer. The presence of pathologic mutations can guide patient selection for breast conserving surgery vs mastectomy. However, our understanding of the interplay between genetic variants and radiotherapy responses and side effects remains incomplete. Here we review the available data on germline mutations and polymorphisms in breast cancer. We also outline their association with treatment tolerance, locoregional outcomes, and ongoing efforts to transform these insights into more effective treatment strategies in combination with radiotherapy.
Collapse
Affiliation(s)
- Michael D Green
- Department of Radiation Oncology, University of Michigan School of Medicine, Ann Arbor, MI; Rogel Cancer Center, University of Michigan, Ann Arbor, MI; Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, MI; Graduate Program in Immunology, University of Michigan School of Medicine, Ann Arbor, MI; Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI
| | - Randall Brenneman
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO
| | - Simon N Powell
- Department of Radiation Oncology and Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Carmen Bergom
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO.
| |
Collapse
|
2
|
Huang A, Yao J, Liu T, Lin Z, Zhang S, Zhang T, Ma H. 53BP1 loss suppresses the radiosensitizing effect of icotinib hydrochloride in colorectal cancer cells. Int J Radiat Biol 2018; 94:327-334. [PMID: 29388453 DOI: 10.1080/09553002.2018.1434322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND This study aimed to investigate the influence of the expression of P53-binding protein 1 (53BP1), a key component in DNA damage repair pathways, on the radiosensitizing effect of icotinib hydrochloride in colorectal cancer and to elucidate the mechanisms underlying this influence. MATERIALS AND METHODS Real-time RT-PCR and Western blotting were performed to verify the gene-knockout effect of 53BP1 small hairpin RNA (ShRNA), and colony formation assay was employed to investigate the influence of 53BP1 downregulation on the radiosensitizing effect of icotinib hydrochloride in HCT116 cells. Cell apoptosis, cell cycle distributions, and histone H2AX (γ-H2AX) fluorescence foci after 53BP1 knockdown were evaluated. Relative protein expression in the ataxia telangiectasia mutated kinase (ATM)-checkpoint kinase-2 (CHK2)-P53 pathway was measured by Western blot analysis to unravel the molecular mechanisms linking the pathway to the above phenomena. RESULTS Icotinib hydrochloride increased the radiosensitivity of HCT116 cells; however, this effect was suppressed by the downregulation of 53BP1 expression, a change that inhibited cell apoptosis, increased the percentage of HCT116 cells arrested in S-phase and inhibited the protein expression of key molecules in the ATM-CHK2-P53 apoptotic pathway. CONCLUSION Our studies confirmed that the loss of 53BP1 serves as a negative regulator of the radiosensitizing effect of icotinib in part by suppressing the ATM-CHK2-P53 apoptotic pathway.
Collapse
Affiliation(s)
- Ai Huang
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Jing Yao
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Tao Liu
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Zhenyu Lin
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Sheng Zhang
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Tao Zhang
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Hong Ma
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| |
Collapse
|
3
|
Yao J, Huang A, Zheng X, Liu T, Lin Z, Zhang S, Yang Q, Zhang T, Ma H. 53BP1 loss induces chemoresistance of colorectal cancer cells to 5-fluorouracil by inhibiting the ATM-CHK2-P53 pathway. J Cancer Res Clin Oncol 2016; 143:419-431. [PMID: 27838786 DOI: 10.1007/s00432-016-2302-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/05/2016] [Indexed: 01/23/2023]
Abstract
PURPOSE Loss of P53 binding protein 1 (53BP1) is considered a poor prognostic factor for colorectal cancer. However, its effect on chemosensitivity of colorectal cancer to 5-fluorouracil (5-FU) remains elusive. This study aimed to examine the association of 53BP1 expression with chemosensitivity of colorectal cancer cells to 5-FU. METHODS Immunohistochemistry was performed on 30 metastatic colorectal cancer samples to assess the associations of 53BP1 levels with clinical therapeutic effects. In vitro, IC50 values for 5-FU and 53BP1 levels were determined by MTT assay and Western blot in 5 colorectal cancer cell lines. Then, 53BP1 was silenced in HCT116 and HT29 cells, and cell proliferation, apoptosis and cell cycle distribution were evaluated. Relative protein levels of ATM-CHK2-P53 pathway effectors and Bcl-2 family members were measured by Western blot. Finally, the effects of 53BP1 knockdown on tumor growth and 5-FU chemoresistance were investigated in vivo. RESULTS 53BP1 expression was closely related to time to progression (TTP) after first-line chemotherapy. Namely, 53BP1 downregulation resulted in reduced TTP. In addition, 53BP1 silencing increased proliferation, inhibited apoptosis and induced S phase arrest in HCT116 and HT29 cells after 5-FU treatment. Moreover, 53BP1 knockdown also reduced the protein levels of ATM-CHK2-P53 apoptotic pathway effectors, caspase9 and caspase3, while increasing Bcl-2 expression. In vivo, 53BP1 silencing accelerated tumor proliferation in nude mice and enhanced resistance to 5-FU. CONCLUSIONS These findings confirmed that 53BP1 loss might be a negative factor for chemotherapy efficacy, promoting cell proliferation and inhibiting apoptosis by suppressing ATM-CHK2-P53 signaling, and finally inducing 5-FU resistance.
Collapse
Affiliation(s)
- Jing Yao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, Hubei, China
| | - Ai Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, Hubei, China
| | - Xiumei Zheng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, Hubei, China
| | - Tao Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, Hubei, China
| | - Zhenyu Lin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, Hubei, China
| | - Sheng Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, Hubei, China
| | - Qin Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, Hubei, China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, Hubei, China
| | - Hong Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, Hubei, China.
| |
Collapse
|
4
|
Liu L, Jiao J, Wang Y, Zhang D, Wu J, Huang D. Lack of association of the TP53BP1 Glu353Asp polymorphism with risk of cancer: a systematic review and meta-analysis. PLoS One 2014; 9:e90931. [PMID: 24603722 PMCID: PMC3946247 DOI: 10.1371/journal.pone.0090931] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 02/04/2014] [Indexed: 11/19/2022] Open
Abstract
Objective The TP53BP1 gene may be involved in the development of cancer through disrupting DNA repair. However, studies investigating the relationship between TP53BP1 Glu353Asp (rs560191) polymorphism and cancer yielded contradictory and inconclusive outcomes. In order to realize these ambiguous findings, a meta-analysis was performed to assess the association between the TP53BP1 Glu353Asp (rs560191) polymorphism and susceptibility to cancer. Methods We conducted a search of all English reports on studies for the association between the TP53BP1 Asp353Glu (rs560191) polymorphism and susceptibility to cancer using Medline, the Cochrane Library, EMbase, Web of Science, Google (scholar), and all Chinese reports were identified manually and on-line using CBMDisc, Chongqing VIP database, and CNKI database. The strict selection criteria and exclusion criteria were determined, and odds ratios (ORs) with 95% confidence intervals (CIs) were used to assess the strength of associations. The fixed or random effect model was selected based on the heterogeneity test among studies. Publication bias was estimated using funnel plots and Egger’s regression test. Results A total of seven studies were included in the meta-analysis including 3,213 cases and 3,849 controls. The results indicated that the Glu353Asp (rs560191) polymorphism in TP53BP1 gene had no association with cancer risk for all genetic models. In the subgroup analysis, the results suggested that Glu353Asp polymorphism was not associated with the risk of cancer according to ethnicity, cancer type, genotyping method, adjusted with control or not, HWE and quality score. Conclusions This meta-analysis suggested that the Glu353Asp (rs560191) polymorphism in TP53BP1 gene was not associated with risk of cancer.
Collapse
Affiliation(s)
- Lei Liu
- Department of Ophthalmology, The First Affiliated Hospital, China Medical University, Shenyang City, Liaoning Province, China
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang City, Liaoning Province, China
- * E-mail:
| | - Jinghua Jiao
- Department of Anesthesiology, Fengtian Hospital, Shenyang Medical College, Shenyang City, Liaoning Province, China
| | - Yu Wang
- Department of Development and Planning office, China Medical University, Shenyang City, Liaoning Province, China
| | - Dong Zhang
- Department of General Surgery, the Fourth People’s Hospital of Shenyang, Shenyang City, Liaoning Province, China
| | - Jingyang Wu
- Department of Ophthalmology, The First Affiliated Hospital, China Medical University, Shenyang City, Liaoning Province, China
| | - Desheng Huang
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang City, Liaoning Province, China
| |
Collapse
|
5
|
Mehta MS, Dolfi SC, Bronfenbrener R, Bilal E, Chen C, Moore D, Lin Y, Rahim H, Aisner S, Kersellius RD, Teh J, Chen S, Toppmeyer DL, Medina DJ, Ganesan S, Vazquez A, Hirshfield KM. Metabotropic glutamate receptor 1 expression and its polymorphic variants associate with breast cancer phenotypes. PLoS One 2013; 8:e69851. [PMID: 23922822 PMCID: PMC3724883 DOI: 10.1371/journal.pone.0069851] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 06/12/2013] [Indexed: 12/22/2022] Open
Abstract
Several epidemiological studies have suggested a link between melanoma and breast cancer. Metabotropic glutamate receptor 1 (GRM1), which is involved in many cellular processes including proliferation and differentiation, has been implicated in melanomagenesis, with ectopic expression of GRM1 causing malignant transformation of melanocytes. This study was undertaken to evaluate GRM1 expression and polymorphic variants in GRM1 for associations with breast cancer phenotypes. Three single nucleotide polymorphisms (SNPs) in GRM1 were evaluated for associations with breast cancer clinicopathologic variables. GRM1 expression was evaluated in human normal and cancerous breast tissue and for in vitro response to hormonal manipulation. Genotyping was performed on genomic DNA from over 1,000 breast cancer patients. Rs6923492 and rs362962 genotypes associated with age at diagnosis that was highly dependent upon the breast cancer molecular phenotype. The rs362962 TT genotype also associated with risk of estrogen receptor or progesterone receptor positive breast cancer. In vitro analysis showed increased GRM1 expression in breast cancer cells treated with estrogen or the combination of estrogen and progesterone, but reduced GRM1 expression with tamoxifen treatment. Evaluation of GRM1 expression in human breast tumor specimens demonstrated significant correlations between GRM1 staining with tissue type and molecular features. Furthermore, analysis of gene expression data from primary breast tumors showed that high GRM1 expression correlated with a shorter distant metastasis-free survival as compared to low GRM1 expression in tamoxifen-treated patients. Additionally, induced knockdown of GRM1 in an estrogen receptor positive breast cancer cell line correlated with reduced cell proliferation. Taken together, these findings suggest a functional role for GRM1 in breast cancer.
Collapse
Affiliation(s)
- Madhura S. Mehta
- Division of Medical Oncology, Department of Medicine, The Cancer Institute of New Jersey/University of Medicine and Dentistry of New Jersey - Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Sonia C. Dolfi
- Division of Medical Oncology, Department of Medicine, The Cancer Institute of New Jersey/University of Medicine and Dentistry of New Jersey - Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Roman Bronfenbrener
- Division of Medical Oncology, Department of Medicine, The Cancer Institute of New Jersey/University of Medicine and Dentistry of New Jersey - Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Erhan Bilal
- Division of Medical Oncology, Department of Medicine, The Cancer Institute of New Jersey/University of Medicine and Dentistry of New Jersey - Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Chunxia Chen
- Department of Biometrics, The Cancer Institute of New Jersey/University of Medicine and Dentistry of New Jersey - Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Dirk Moore
- Department of Biometrics, The Cancer Institute of New Jersey/University of Medicine and Dentistry of New Jersey - Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Yong Lin
- Department of Biometrics, The Cancer Institute of New Jersey/University of Medicine and Dentistry of New Jersey - Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Hussein Rahim
- Division of Medical Oncology, Department of Medicine, The Cancer Institute of New Jersey/University of Medicine and Dentistry of New Jersey - Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Seena Aisner
- Department of Pathology and Laboratory Medicine, University of Medicine and Dentistry of New Jersey - New Jersey Medical School, Newark, New Jersey, United States of America
| | - Romona D. Kersellius
- Division of Medical Oncology, Department of Medicine, The Cancer Institute of New Jersey/University of Medicine and Dentistry of New Jersey - Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Jessica Teh
- Department of Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Suzie Chen
- Department of Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Deborah L. Toppmeyer
- Division of Medical Oncology, Department of Medicine, The Cancer Institute of New Jersey/University of Medicine and Dentistry of New Jersey - Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Dan J. Medina
- Division of Medical Oncology, Department of Medicine, The Cancer Institute of New Jersey/University of Medicine and Dentistry of New Jersey - Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Shridar Ganesan
- Division of Medical Oncology, Department of Medicine, The Cancer Institute of New Jersey/University of Medicine and Dentistry of New Jersey - Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Alexei Vazquez
- Department of Radiation Oncology, The Cancer Institute of New Jersey/University of Medicine and Dentistry of New Jersey- Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Kim M. Hirshfield
- Division of Medical Oncology, Department of Medicine, The Cancer Institute of New Jersey/University of Medicine and Dentistry of New Jersey - Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
6
|
Li X, Xu B, Moran MS, Zhao Y, Su P, Haffty BG, Shao C, Yang Q. 53BP1 functions as a tumor suppressor in breast cancer via the inhibition of NF-κB through miR-146a. Carcinogenesis 2012; 33:2593-600. [PMID: 23027628 DOI: 10.1093/carcin/bgs298] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
p53-binding protein-1 (53BP1) plays a critical role in cell cycle checkpoint and DNA repair activities. Recently, 53BP1 was recognized as a potential tumor suppressor gene. In this study, we investigated its tumor suppressor function in breast cancer. In clinical samples, we observed a lower level of 53BP1 expression in the cancer lesions than in the matched non-tumor tissues. Furthermore, the 53BP1 level showed a gradual decrease during the progression from precancerous to cancer lesion. Ectopic expression of 53BP1 can significantly inhibit cell proliferation and curb the invasiveness in breast cancer cell lines, whereas knockdown of 53BP1 by RNA interference had the opposite effects. Additionally, 53BP1 markedly inhibited xenograft formation and metastasis of breast cancer cells in nude mice. Both in vitro and in vivo studies revealed that the 53BP1 expression level was inversely correlated to the function of nuclear factor-kappaB (NF-κB), which contributes to the invasion and metastasis of breast cancer. Importantly, the inhibitory effect of 53BP1 on NF-κB activity was shown to be mediated by the upregulation of miR-146a. Together, our findings demonstrated that 53BP1 has a potent tumor suppressor activity in breast cancer, and it may serve as a novel target for breast cancer prevention and treatment.
Collapse
Affiliation(s)
- Xiaoyan Li
- Department of Breast Surgery, Qilu Hospital, Shandong University, Wenhua Xi Road No.107, Jinan 250012, China
| | | | | | | | | | | | | | | |
Collapse
|
7
|
The impact of pharmacogenetics on radiation therapy outcome in cancer patients. A focus on DNA damage response genes. Cancer Treat Rev 2012; 38:737-59. [DOI: 10.1016/j.ctrv.2012.02.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 02/03/2012] [Accepted: 02/06/2012] [Indexed: 11/18/2022]
|
8
|
Neboori HJR, Haffty BG, Wu H, Yang Q, Aly A, Goyal S, Schiff D, Moran MS, Golhar R, Chen C, Moore D, Ganesan S. Low p53 binding protein 1 (53BP1) expression is associated with increased local recurrence in breast cancer patients treated with breast-conserving surgery and radiotherapy. Int J Radiat Oncol Biol Phys 2012; 83:e677-83. [PMID: 22520477 DOI: 10.1016/j.ijrobp.2012.01.089] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 01/27/2012] [Accepted: 01/31/2012] [Indexed: 11/24/2022]
Abstract
PURPOSE To investigate whether the expression of p53 binding protein 1 (53BP1) has prognostic significance in a cohort of early-stage breast cancer patients treated with breast-conserving surgery and radiotherapy (BCS+RT). METHODS AND MATERIALS A tissue microarray of early-stage breast cancer treated with BCS+RT from a cohort of 514 women was assayed for 53BP1, estrogen receptor, progesterone receptor, and HER2 expression by immunohistochemistry. Through log-rank tests and univariate and multivariate models, the staining profile of each tumor was correlated with clinical endpoints, including ipsilateral breast recurrence-free survival (IBRFS), distant metastasis-free survival (DMFS), cause-specific survival (CSS), recurrence-free survival (RFS), and overall survival (OS). RESULTS Of the 477 (93%) evaluable tumors, 63 (13%) were scored as low. Low expression of 53BP1 was associated with worse outcomes for all endpoints studied, including 10-year IBRFS (76.8% vs. 90.5%; P=.01), OS (66.4% vs. 81.7%; P=.02), CSS (66.0% vs. 87.4%; P<.01), DMFS (55.9% vs. 87.0%; P<.01), and RFS (45.2% vs. 80.6%; P<.01). Multivariate analysis incorporating various clinico-pathologic markers and 53BP1 expression found that 53BP1 expression was again an independent predictor of all endpoints (IBRFS: P=.0254; OS: P=.0094; CSS: P=.0033; DMFS: P=.0006; RFS: P=.0002). Low 53BP1 expression was also found to correlate with triple-negative (TN) phenotype (P<.01). Furthermore, in subset analysis of all TN breast cancer, negative 53BP1 expression trended for lower IBRFS (72.3% vs. 93.9%; P=.0361) and was significant for worse DMFS (48.2% vs. 86.8%; P=.0035) and RFS (37.8% vs. 83.7%; P=.0014). CONCLUSION Our data indicate that low 53BP1 expression is an independent prognostic indicator for local relapse among other endpoints in early-stage breast cancer and TN breast cancer patients treated with BCS+RT. These results should be verified in larger cohorts of patients to validate their clinical significance.
Collapse
Affiliation(s)
- Hanmanth J R Neboori
- Department of Radiation Oncology, The Cancer Institute of New Jersey and University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|