1
|
Qiu Z, Sigh D, Liu Y, Prasad CB, Bean N, Yan C, Li Z, Zhang X, Narla G, DiFeo A, Wang QE, Zhang J. Low PPP2R2A expression promotes sensitivity to CHK1 inhibition in high-grade serous ovarian cancer. Theranostics 2024; 14:7450-7469. [PMID: 39659585 PMCID: PMC11626944 DOI: 10.7150/thno.96879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/06/2024] [Indexed: 12/12/2024] Open
Abstract
Rationale: High-grade serous ovarian cancer (HGSOC), the most lethal epithelial ovarian cancer subtype, faces persistent challenges despite advances in the therapeutic use of PARP inhibitors. Thus, innovative strategies are urgently needed to improve survival rates for this deadly disease. Checkpoint kinase 1 (CHK1) is pivotal in regulating cell survival during oncogene-induced replication stress (RS). While CHK1 inhibitors (CHK1i's) show promise as monotherapy for ovarian cancer, a crucial biomarker for effective stratification in clinical trials is lacking, hindering efficacy improvement and toxicity reduction. PP2A B55α, encoded by PPP2R2A, is a regulatory subunit of the serine/threonine protein phosphatase 2 (PP2A) that influences CHK1 sensitivity in non-small cell lung cancer (NSCLC). Given the complexity of PP2A B55α function in different types of cancer, here we sought to identify whether PPP2R2A deficiency enhances the sensitivity of HGSOC to CHK1 inhibition. Methods: To determine whether PPP2R2A deficiency affects the sensitivity of HGSOC to CHK1 inhibition, we treated PPP2R2A knockdown (KD) HGSOC cells or HGSOC cells with naturally low PPP2R2A expression with a CHK1 inhibitor, then assessed cell growth in in vitro and in vivo assays. Additionally, we investigated the mechanisms contributing to the increased RS and the enhanced sensitivity to the CHK1 inhibitor in PPP2R2A-KD or deficient cells using various molecular biology assays, including western blotting, immunofluorescence, and DNA fiber assays. Results: Our study suggests that PPP2R2A-KD elevates c-Myc-induced RS via upregulation of replication initiation, rendering HGSOC cells reliant on CHK1 for survival, including those resistant to PARP inhibitors. Conclusion: Combined, these results identify PPP2R2A/PP2A B55α as a potential predictive biomarker for CHK1i sensitivity in HGSOC, as well as suggesting it as a therapeutic target to overcome PARP resistance.
Collapse
Affiliation(s)
- Zhaojun Qiu
- Department of Radiation Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio-43210, United States
| | - Deepika Sigh
- Department of Radiation Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio-43210, United States
| | - Yujie Liu
- Department of Radiation Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio-43210, United States
| | - Chandra B. Prasad
- Department of Radiation Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio-43210, United States
| | - Nichalos Bean
- Department of Radiation Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio-43210, United States
| | - Chunhong Yan
- Georgia Cancer Center, Augusta University Medical College, 1410 Laney Walker Blvd., CN-2134, Augusta, Georgia-30912, United States
| | - Zaibo Li
- Department of Pathology, The Ohio State University Wexner Medical Center, College of Medicine, Columbus, Ohio-43210, United States
| | - Xiaoli Zhang
- Department of Biomedical Informatics, Wexner Medical Center, College of Medicine, The Ohio State University, Ohio-43210, United States
| | - Goutham Narla
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI-48109, United States
| | - Analisa DiFeo
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI-48109, United States
| | - Qi-En Wang
- Department of Radiation Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio-43210, United States
| | - Junran Zhang
- Department of Radiation Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio-43210, United States
- The James Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, Ohio-43210, United States
- The James Comprehensive Cancer Center, Center for metabolism, The Ohio State University, Columbus, Ohio-43210, United States
| |
Collapse
|
2
|
Melia E, Parsons JL. The Potential for Targeting G 2/M Cell Cycle Checkpoint Kinases in Enhancing the Efficacy of Radiotherapy. Cancers (Basel) 2024; 16:3016. [PMID: 39272874 PMCID: PMC11394570 DOI: 10.3390/cancers16173016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Radiotherapy is one of the main cancer treatments being used for ~50% of all cancer patients. Conventional radiotherapy typically utilises X-rays (photons); however, there is increasing use of particle beam therapy (PBT), such as protons and carbon ions. This is because PBT elicits significant benefits through more precise dose delivery to the cancer than X-rays, but also due to the increases in linear energy transfer (LET) that lead to more enhanced biological effectiveness. Despite the radiotherapy type, the introduction of DNA damage ultimately drives the therapeutic response through stimulating cancer cell death. To combat this, cells harbour cell cycle checkpoints that enables time for efficient DNA damage repair. Interestingly, cancer cells frequently have mutations in key genes such as TP53 and ATM that drive the G1/S checkpoint, whereas the G2/M checkpoint driven through ATR, Chk1 and Wee1 remains intact. Therefore, targeting the G2/M checkpoint through specific inhibitors is considered an important strategy for enhancing the efficacy of radiotherapy. In this review, we focus on inhibitors of Chk1 and Wee1 kinases and present the current biological evidence supporting their utility as radiosensitisers with different radiotherapy modalities, as well as clinical trials that have and are investigating their potential for cancer patient benefit.
Collapse
Affiliation(s)
- Emma Melia
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Jason L Parsons
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
- School of Physics and Astronomy, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
3
|
Prasad CB, Oo A, Liu Y, Qiu Z, Zhong Y, Li N, Singh D, Xin X, Cho YJ, Li Z, Zhang X, Yan C, Zheng Q, Wang QE, Guo D, Kim B, Zhang J. The thioredoxin system determines CHK1 inhibitor sensitivity via redox-mediated regulation of ribonucleotide reductase activity. Nat Commun 2024; 15:4667. [PMID: 38821952 PMCID: PMC11143221 DOI: 10.1038/s41467-024-48076-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 04/19/2024] [Indexed: 06/02/2024] Open
Abstract
Checkpoint kinase 1 (CHK1) is critical for cell survival under replication stress (RS). CHK1 inhibitors (CHK1i's) in combination with chemotherapy have shown promising results in preclinical studies but have displayed minimal efficacy with substantial toxicity in clinical trials. To explore combinatorial strategies that can overcome these limitations, we perform an unbiased high-throughput screen in a non-small cell lung cancer (NSCLC) cell line and identify thioredoxin1 (Trx1), a major component of the mammalian antioxidant-system, as a determinant of CHK1i sensitivity. We establish a role for redox recycling of RRM1, the larger subunit of ribonucleotide reductase (RNR), and a depletion of the deoxynucleotide pool in this Trx1-mediated CHK1i sensitivity. Further, the TrxR inhibitor auranofin, an approved anti-rheumatoid arthritis drug, shows a synergistic interaction with CHK1i via interruption of the deoxynucleotide pool. Together, we show a pharmacological combination to treat NSCLC that relies on a redox regulatory link between the Trx system and mammalian RNR activity.
Collapse
Affiliation(s)
- Chandra Bhushan Prasad
- Department of Radiation Oncology, James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Adrian Oo
- Center for ViroScience and Cure, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Yujie Liu
- Department of Radiation Oncology, James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Zhaojun Qiu
- Department of Radiation Oncology, James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Yaogang Zhong
- Department of Radiation Oncology, James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA
- The Comprehensive Cancer Center, Center for Cancer Metabolism, The Ohio State University, Columbus, OH, 43210, USA
| | - Na Li
- Department of Radiation Oncology, James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Deepika Singh
- Department of Radiation Oncology, James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Xiwen Xin
- The Ohio State University, Columbus, OH, 43210, USA
| | - Young-Jae Cho
- Center for ViroScience and Cure, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Zaibo Li
- Department of Pathology, The Ohio State University Wexner Medical Center, College of Medicine, Columbus, OH, 43210, USA
| | - Xiaoli Zhang
- Department of Biomedical Informatics, Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Chunhong Yan
- Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA
| | - Qingfei Zheng
- Department of Radiation Oncology, James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA
- The Comprehensive Cancer Center, Center for Cancer Metabolism, The Ohio State University, Columbus, OH, 43210, USA
| | - Qi-En Wang
- Department of Radiation Oncology, James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Deliang Guo
- Department of Radiation Oncology, James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA
- The Comprehensive Cancer Center, Center for Cancer Metabolism, The Ohio State University, Columbus, OH, 43210, USA
| | - Baek Kim
- Center for ViroScience and Cure, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Junran Zhang
- Department of Radiation Oncology, James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA.
- The Comprehensive Cancer Center, Center for Cancer Metabolism, The Ohio State University, Columbus, OH, 43210, USA.
- The Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
4
|
Xie D, Huang Q, Zhou P. Drug Discovery Targeting Post-Translational Modifications in Response to DNA Damages Induced by Space Radiation. Int J Mol Sci 2023; 24:ijms24087656. [PMID: 37108815 PMCID: PMC10142602 DOI: 10.3390/ijms24087656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/07/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
DNA damage in astronauts induced by cosmic radiation poses a major barrier to human space exploration. Cellular responses and repair of the most lethal DNA double-strand breaks (DSBs) are crucial for genomic integrity and cell survival. Post-translational modifications (PTMs), including phosphorylation, ubiquitylation, and SUMOylation, are among the regulatory factors modulating a delicate balance and choice between predominant DSB repair pathways, such as non-homologous end joining (NHEJ) and homologous recombination (HR). In this review, we focused on the engagement of proteins in the DNA damage response (DDR) modulated by phosphorylation and ubiquitylation, including ATM, DNA-PKcs, CtIP, MDM2, and ubiquitin ligases. The involvement and function of acetylation, methylation, PARylation, and their essential proteins were also investigated, providing a repository of candidate targets for DDR regulators. However, there is a lack of radioprotectors in spite of their consideration in the discovery of radiosensitizers. We proposed new perspectives for the research and development of future agents against space radiation by the systematic integration and utilization of evolutionary strategies, including multi-omics analyses, rational computing methods, drug repositioning, and combinations of drugs and targets, which may facilitate the use of radioprotectors in practical applications in human space exploration to combat fatal radiation hazards.
Collapse
Affiliation(s)
- Dafei Xie
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Taiping Road 27th, Haidian District, Beijing 100850, China
| | - Qi Huang
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Taiping Road 27th, Haidian District, Beijing 100850, China
- Department of Preventive Medicine, School of Public Health, University of South China, Changsheng West Road 28th, Zhengxiang District, Hengyang 421001, China
| | - Pingkun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Taiping Road 27th, Haidian District, Beijing 100850, China
- Department of Preventive Medicine, School of Public Health, University of South China, Changsheng West Road 28th, Zhengxiang District, Hengyang 421001, China
| |
Collapse
|
5
|
Toulany M. Targeting K-Ras-mediated DNA damage response in radiation oncology: Current status, challenges and future perspectives. Clin Transl Radiat Oncol 2022; 38:6-14. [PMID: 36313934 PMCID: PMC9596599 DOI: 10.1016/j.ctro.2022.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
Abstract
Approximately 60% of cancer patients receive curative or palliative radiation. Despite the significant role of radiotherapy (RT) as a curative approach for many solid tumors, tumor recurrence occurs, partially because of intrinsic radioresistance. Accumulating evidence indicates that the success of RT is hampered by activation of the DNA damage response (DDR). The intensity of DDR signaling is affected by multiple parameters, e.g., loss-of-function mutations in tumor suppressor genes, gain-of-function mutations in protooncogenes as well as radiation-induced alterations in signal-transduction pathways. Therefore, the response to irradiation differs in tumors of different types, which makes the individualization of RT as a rational but challenging goal. One contributor to tumor cell radiation survival is signaling through the Ras pathway. Three RAS genes encode 4 Ras isoforms: K-Ras4A, K-Ras4B, H-Ras, and N-Ras. RAS family members are found to be mutated in approximately 19% of human cancers. Mutations in RAS lead to constitutive activation of the gene product and activation of multiple Ras-dependent signal-transduction cascades. Preclinical studies have shown that the expression of mutant KRAS affects DDR and increases cell survival after irradiation. Approximately 70% of RAS mutations occur in KRAS. Thus, applying targeted therapies directly against K-Ras as well as K-Ras upstream activators and downstream effectors might be a tumor-specific approach to overcome K-Ras-mediated RT resistance. In this review, the role of K-Ras in the activation of DDR signaling will be summarized. Recent progress in targeting DDR in KRAS-mutated tumors in combination with radiochemotherapy will be discussed.
Collapse
|
6
|
Ozgiray E, Sogutlu F, Biray Avci C. Chk1/2 inhibitor AZD7762 enhances the susceptibility of IDH-mutant brain cancer cells to temozolomide. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:166. [PMID: 35972603 DOI: 10.1007/s12032-022-01769-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/13/2022] [Indexed: 11/28/2022]
Abstract
The IDH mutation initially exhibits chemosensitive properties, progression-free survival cannot be achieved in the later grades, and malignant transformation occurs as a result of TMZ-induced hypermutation profile and adaptation to this profile. In this study, we evaluated the potential of the combination of TMZ and AZD7762 at molecular level, to increase the anticancer activity of TMZ in IDH-mutant U87-mg cells. We used the WST-1 test to evaluate cytotoxic effect of TMZ and AZD7762 combination with dose-effect and isobologram curves. The effects of the inhibitory and effective concentrations of the combination on apoptosis, cell cycle and γ-H2AX phosphorylation were analyzed with flow cytometry. The expression of genes responsible for the DNA damage response was analyzed with qRT-PCR. The combination showed a synergistic effect with high dose reduction index. Single and combined administrations of TMZ and AZD7762 increased in G2/M arrest from 24 to 48 h, and cells in the G2/M phase shifted towards octaploidy at 72 h. While no double-strand breaks were detected after TMZ treatment, AZD7762 and combination treatments caused a significant increase in γ-H2AX phosphorylation and increased apoptotic stimulation towards 72 h although TMZ did not cause apoptotic effect in IDH-mutant U87-mg cells. The genes controlling the apoptosis were determined to be upregulated in all three groups, and genes regarding cell cycle checkpoints were downregulated. Targeting Chk1/2 with AZD7762 simultaneously with TMZ may be a potential therapeutic strategy for both increasing the sensitivity of IDH-mutant glioma cells to TMZ and reducing the dose of TMZ. In IDH-mutant glioma cells, AZD7762, the Chk1/2 inhibitor, can increase the efficacy of Temozolomide by (i) increasing mitotic chaos, and (ii) inhibiting double-strand break repair, (iii) thereby inducing cell death.
Collapse
Affiliation(s)
- Erkin Ozgiray
- Department of Neurosurgery, Medicine Faculty, Ege University, Izmir, Turkey
| | - Fatma Sogutlu
- Department of Medical Biology, Medicine Faculty, Ege University, Izmir, Turkey
| | - Cigir Biray Avci
- Department of Medical Biology, Medicine Faculty, Ege University, Izmir, Turkey.
| |
Collapse
|
7
|
An J, Peng C, Xie X, Peng F. New Advances in Targeted Therapy of HER2-Negative Breast Cancer. Front Oncol 2022; 12:828438. [PMID: 35311116 PMCID: PMC8931202 DOI: 10.3389/fonc.2022.828438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/10/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer has an extremely high incidence in women, and its morbidity and mortality rank first among female tumors. With the increasing development of molecular biology and genomics, molecular targeted therapy has become one of the most active areas in breast cancer treatment research and has also achieved remarkable achievements. However, molecular targeted therapy is mainly aimed at HER2-positive breast cancer and has not yet achieved satisfactory curative effect on HER2-negative breast cancer. This article describes the potential targets that may be used for breast cancer treatment from the aspects of PI3K/AKT signaling pathway, DDR, angiogenesis, the cell cycle, breast cancer stem cells, etc., and explores possible inhibitors for the treatment of HER2-negative breast cancer, such as PI3K inhibitors, AKT inhibitors and m-TOR inhibitors that inhibit the PI3K/AKT signaling pathway, small molecule tyrosine kinase inhibitors that restrain angiogenesis, CDK inhibitors, aurora kinase inhibitors and HDAC inhibitors that block cell cycle, as well as the drugs targeting breast cancer stem cells which have been a hit, aiming to provide a new idea and strategy for the treatment of HER2-negative breast cancer.
Collapse
Affiliation(s)
- Junsha An
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Cheng Peng
- State Key Laboratory Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Xie
- State Key Laboratory Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fu Peng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
- State Key Laboratory Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
8
|
Casey DL, Pitter KL, Wexler LH, Slotkin EK, Gupta GP, Wolden SL. TP53 mutations increase radioresistance in rhabdomyosarcoma and Ewing sarcoma. Br J Cancer 2021; 125:576-581. [PMID: 34017087 PMCID: PMC8368014 DOI: 10.1038/s41416-021-01438-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/28/2021] [Accepted: 05/10/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND p53 plays a key role in the DNA repair process and response to ionising radiation. We sought to determine the clinical phenotype of TP53 mutations and p53 pathway alterations in patients with rhabdomyosarcoma (RMS) and Ewing sarcoma (ES) treated with radiation. METHODS Of patients with available genomic sequencing, we identified 109 patients with RMS and ES treated to a total of 286 radiation sites. We compared irradiated tumour control among tumours with TP53 mutations (n = 40) to those that were TP53 wild-type (n = 246). We additionally compared irradiated tumour control among tumours with any p53 pathway alteration (defined as tumours with TP53 mutations or TP53 wild-type tumours identified to have MDM2/4 amplification and/or CDKN2A/B deletion, n = 78) to those without such alterations (n = 208). RESULTS The median follow-up was 26 months from radiation. TP53 mutations were associated with worse irradiated tumour control among the entire cohort (hazard ratio, HR = 2.8, P < 0.0001). Tumours with any p53 pathway alteration also had inferior irradiated tumour control (HR = 2.0, P = 0.003). On multivariable analysis, after controlling for tumour histology, intent of radiation, presence of gross disease, and biologically effective dose, TP53 mutations continued to be associated with a radioresistant phenotype (HR = 7.1, P < 0.0001). CONCLUSIONS Our results show that TP53 mutations are associated with increased radioresistance in RMS and ES. Novel strategies to overcome this radioresistance are important for improved outcomes in p53 disruptive RMS and ES.
Collapse
Affiliation(s)
- Dana L. Casey
- grid.51462.340000 0001 2171 9952Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY USA ,grid.10698.360000000122483208Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, NC USA ,grid.429995.aLineberger Comprehensive Cancer Center, University of North Carolina Hospitals, Chapel Hill, NC USA
| | - Kenneth L. Pitter
- grid.51462.340000 0001 2171 9952Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Leonard H. Wexler
- grid.51462.340000 0001 2171 9952Department of Pediatric Oncology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Emily K. Slotkin
- grid.51462.340000 0001 2171 9952Department of Pediatric Oncology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Gaorav P. Gupta
- grid.10698.360000000122483208Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, NC USA ,grid.429995.aLineberger Comprehensive Cancer Center, University of North Carolina Hospitals, Chapel Hill, NC USA
| | - Suzanne L. Wolden
- grid.51462.340000 0001 2171 9952Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| |
Collapse
|
9
|
Inhibition of the DSB repair protein RAD51 potentiates the cytotoxic efficacy of doxorubicin via promoting apoptosis-related death pathways. Cancer Lett 2021; 520:361-373. [PMID: 34389435 DOI: 10.1016/j.canlet.2021.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 01/04/2023]
Abstract
The anthracycline derivative doxorubicin (Doxo) induces DNA double-strand breaks (DSBs) by inhibition of DNA topoisomerase type II. Defective mismatch repair (MMR) contributes to Doxo resistance and has been reported for colon and mammary carcinomas. Here, we investigated the outcome of pharmacological inhibition of various DNA repair-related mechanisms on Doxo-induced cytotoxicity employing MMR-deficient HCT-116 colon carcinoma cells. Out of different inhibitors tested (i.e. HDACi, PARPi, MRE11i, RAD52i, RAD51i), we identified the RAD51-inhibitor B02 as the most powerful compound to synergistically increase Doxo-induced cytotoxicity. B02-mediated synergism rests on pleiotropic mechanisms, including pronounced G2/M arrest, damage to mitochondria and caspase-driven apoptosis. Of note, B02 also promotes the cytotoxicity of oxaliplatin and 5-fluoruracil (5-FU) in HCT-116 cells and, furthermore, also increases Doxo-induced cytotoxicity in MMR-proficient colon and mammary carcinoma cells. Summarizing, pharmacological inhibition of RAD51 is suggested to synergistically increase the cytotoxic efficacy of various types of conventional anticancer drugs in different tumor entities. Hence, pre-clinical in vivo studies are preferable to determine the therapeutic window of B02 in a clinically oriented therapeutic regimen.
Collapse
|
10
|
Fernandes SG, Shah P, Khattar E. Recent Advances in Therapeutic Application of DNA Damage Response Inhibitors against Cancer. Anticancer Agents Med Chem 2021; 22:469-484. [PMID: 34102988 DOI: 10.2174/1871520621666210608105735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/02/2021] [Accepted: 02/22/2021] [Indexed: 11/22/2022]
Abstract
DNA integrity is continuously challenged by intrinsic cellular processes and environmental agents. To overcome this genomic damage, cells have developed multiple signaling pathways collectively named as DNA damage response (DDR) and composed of three components: (i) sensor proteins, which detect DNA damage, (ii) mediators that relay the signal downstream and recruit the repair machinery, and (iii) the repair proteins, which restore the damaged DNA. A flawed DDR and failure to repair the damage lead to the accumulation of genetic lesions and increased genomic instability, which is recognized as a hallmark of cancer. Cancer cells tend to harbor increased mutations in DDR genes and often have fewer DDR pathways than normal cells. This makes cancer cells more dependent on particular DDR pathways and thus become more susceptible to compounds inhibiting those pathways compared to normal cells, which have all the DDR pathways intact. Understanding the roles of different DDR proteins in the DNA damage response and repair pathways and identification of their structures have paved the way for the development of their inhibitors as targeted cancer therapy. In this review, we describe the major participants of various DDR pathways, their significance in carcinogenesis, and focus on the inhibitors developed against several key DDR proteins.
Collapse
Affiliation(s)
- Stina George Fernandes
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be) University, Mumbai, India
| | - Prachi Shah
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be) University, Mumbai, India
| | - Ekta Khattar
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be) University, Mumbai, India
| |
Collapse
|
11
|
Pesch AM, Pierce LJ, Speers CW. Modulating the Radiation Response for Improved Outcomes in Breast Cancer. JCO Precis Oncol 2021; 5:PO.20.00297. [PMID: 34250414 DOI: 10.1200/po.20.00297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/12/2020] [Accepted: 12/22/2020] [Indexed: 12/25/2022] Open
Affiliation(s)
- Andrea M Pesch
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI.,Department of Pharmacology, University of Michigan, Ann Arbor, MI.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Lori J Pierce
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Corey W Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| |
Collapse
|
12
|
Dreyer SB, Upstill-Goddard R, Paulus-Hock V, Paris C, Lampraki EM, Dray E, Serrels B, Caligiuri G, Rebus S, Plenker D, Galluzzo Z, Brunton H, Cunningham R, Tesson M, Nourse C, Bailey UM, Jones M, Moran-Jones K, Wright DW, Duthie F, Oien K, Evers L, McKay CJ, McGregor GA, Gulati A, Brough R, Bajrami I, Pettitt S, Dziubinski ML, Candido J, Balkwill F, Barry ST, Grützmann R, Rahib L, Johns A, Pajic M, Froeling FE, Beer P, Musgrove EA, Petersen GM, Ashworth A, Frame MC, Crawford HC, Simeone DM, Lord C, Mukhopadhyay D, Pilarsky C, Tuveson DA, Cooke SL, Jamieson NB, Morton JP, Sansom OJ, Bailey PJ, Biankin AV, Chang DK. Targeting DNA Damage Response and Replication Stress in Pancreatic Cancer. Gastroenterology 2021; 160:362-377.e13. [PMID: 33039466 PMCID: PMC8167930 DOI: 10.1053/j.gastro.2020.09.043] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Continuing recalcitrance to therapy cements pancreatic cancer (PC) as the most lethal malignancy, which is set to become the second leading cause of cancer death in our society. The study aim was to investigate the association between DNA damage response (DDR), replication stress, and novel therapeutic response in PC to develop a biomarker-driven therapeutic strategy targeting DDR and replication stress in PC. METHODS We interrogated the transcriptome, genome, proteome, and functional characteristics of 61 novel PC patient-derived cell lines to define novel therapeutic strategies targeting DDR and replication stress. Validation was done in patient-derived xenografts and human PC organoids. RESULTS Patient-derived cell lines faithfully recapitulate the epithelial component of pancreatic tumors, including previously described molecular subtypes. Biomarkers of DDR deficiency, including a novel signature of homologous recombination deficiency, cosegregates with response to platinum (P < .001) and PARP inhibitor therapy (P < .001) in vitro and in vivo. We generated a novel signature of replication stress that predicts response to ATR (P < .018) and WEE1 inhibitor (P < .029) treatment in both cell lines and human PC organoids. Replication stress was enriched in the squamous subtype of PC (P < .001) but was not associated with DDR deficiency. CONCLUSIONS Replication stress and DDR deficiency are independent of each other, creating opportunities for therapy in DDR-proficient PC and after platinum therapy.
Collapse
Affiliation(s)
- Stephan B. Dreyer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom,West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Rosie Upstill-Goddard
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | | | - Clara Paris
- Department of Pharmacological Faculty, Université Grenoble Alpes, Saint-Martin-d’Heres, France
| | - Eirini-Maria Lampraki
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Eloise Dray
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
| | - Bryan Serrels
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom,Medical Research Council Institute of Genetics and Molecular Medicine, Edinburgh Cancer Research UK Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Giuseppina Caligiuri
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Selma Rebus
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Dennis Plenker
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Zachary Galluzzo
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Holly Brunton
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Richard Cunningham
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Mathias Tesson
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Craig Nourse
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Ulla-Maja Bailey
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Marc Jones
- Stratified Medicine Scotland, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Kim Moran-Jones
- College of Medicine, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Derek W. Wright
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Fraser Duthie
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom,Department of Pathology, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Karin Oien
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom,Department of Pathology, Queen Elizabeth University Hospital, Glasgow, United Kingdom,Greater Glasgow and Clyde Bio-repository, Pathology Department, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Lisa Evers
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Colin J. McKay
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom,West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | | | - Aditi Gulati
- Cancer Research UK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Rachel Brough
- Cancer Research UK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Ilirjana Bajrami
- Cancer Research UK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Stephan Pettitt
- Cancer Research UK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Michele L. Dziubinski
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Juliana Candido
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Frances Balkwill
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Simon T. Barry
- Bioscience, Oncology, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, United Kingdom
| | - Robert Grützmann
- Department of Surgery, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lola Rahib
- Pancreatic Cancer Action Network, Manhattan Beach, California
| | - Glasgow Precision Oncology Laboratory,AllisonSarah1BaileyPeter J.1BaileyUlla-Maja1BiankinAndrew V.1BeraldiDario1BruntonHolly1CaligiuriGiuseppina1CameronEuan1ChangDavid K.12CookeSusanna L.1CunninghamRichard1DreyerStephan12GrimwoodPaul1KellyShane1LamprakiEirini-Maria1MarshallJohn1MartinSancha1McDadeBrian1McElroyDaniel1MusgroveElizabeth A.1NourseCraig1Paulus-HockViola1RamsayDonna1Upstill-GoddardRosie1WrightDerek1JonesMarc D.1EversLisa1RebusSelma1RahibLola1SerrelsBryan1HairJane1JamiesonNigel B.12McKayColin J.12WestwoodPaul14WilliamsNicola14DuthieFraser13Glasgow Precision Oncology Laboratory, University of Glasgow, Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, Glasgow, United KingdomWest of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, United KingdomDepartment of Pathology, Southern General Hospital, Greater Glasgow and Clyde National Health Service, Glasgow, United KingdomWest of Scotland Genetic Services, National Health Service, Greater Glasgow and Clyde, Queen Elizabeth University Hospital Campus, Glasgow, United Kingdom
- Glasgow Precision Oncology Laboratory, Glasgow, United Kingdom
| | - Australian Pancreatic Cancer Genome InitiativeBiankinAndrew V.12JohnsAmber L.1MawsonAmanda1ChangDavid K.12ScarlettChristopher J.1BrancatoMary-Anne L.1RoweSarah J.1SimpsonSkye H.1Martyn-SmithMona1ThomasMichelle T.1ChantrillLorraine A.1ChinVenessa T.1ChouAngela1CowleyMark J.1HumphrisJeremy L.1JonesMarc D.12MeadR. Scott1NagrialAdnan M.1PajicMarina1PettitJessica1PineseMark1RoomanIlse1WuJianmin1TaoJiang1DiPietroRenee1WatsonClare1SteinmannAngela1LeeHong Ching1WongRachel1PinhoAndreia V.1Giry-LaterriereMarc1DalyRoger J.1MusgroveElizabeth A.12SutherlandRobert L.1GrimmondSean M.3WaddellNicola3KassahnKarin S.3MillerDavid K.3WilsonPeter J.3PatchAnn-Marie3SongSarah3HarliwongIvon3IdrisogluSenel3NourseCraig3NourbakhshEhsan3ManningSuzanne3WaniShivangi3GongoraMilena3AndersonMatthew3HolmesOliver3LeonardConrad3TaylorDarrin3WoodScott3XuChristina3NonesKatia3FinkJ. Lynn3ChristAngelika3BruxnerTim3CloonanNicole3NewellFelicity3PearsonJohn V.3BaileyPeter3QuinnMichael3NagarajShivashankar3KazakoffStephen3WaddellNick3KrisnanKeerthana3QuekKelly3WoodDavid3SamraJaswinder S.4GillAnthony J.4PavlakisNick4GuminskiAlex4ToonChristopher4AsghariRay5MerrettNeil D.5PaveyDarren5DasAmitabha5CosmanPeter H.6IsmailKasim6O’ConnnorChelsie6LamVincent W.7McLeodDuncan7PleassHenry C.7RichardsonArthur7JamesVirginia7KenchJames G.8CooperCaroline L.8JosephDavid8SandroussiCharbel8CrawfordMichael8GallagherJames8TexlerMichael9ForestCindy9LaycockAndrew9EpariKrishna P.9BallalMo9FletcherDavid R.9MukhedkarSanjay9SpryNigel A.10DeBoerBastiaanChaiMingZepsNikolajs11BeilinMaria11FeeneyKynan11NguyenNan Q.12RuszkiewiczAndrew R.12WorthleyChris12TanChuan P.12DebrenciniTamara12ChenJohn13Brooke-SmithMark E.13PapangelisVirginia13TangHenry14BarbourAndrew P.14CloustonAndrew D.15MartinPatrick15O’RourkeThomas J.16ChiangAmy16FawcettJonathan W.16SlaterKellee16YeungShinn16HatzifotisMichael16HodgkinsonPeter16ChristophiChristopher17NikfarjamMehrdad17MountainAngela17BiobankVictorian Cancer18EshlemanJames R.19HrubanRalph H.19MaitraAnirban19Iacobuzio-DonahueChristine A.19SchulickRichard D.19WolfgangChristopher L.19MorganRichard A.19HodginMary19ScarpaAldo20LawlorRita T.20BeghelliStefania20CorboVincenzo20ScardoniMaria20BassiClaudio20TemperoMargaret A.21BiankinAndrew V.1222GrimmondSean M.23ChangDavid K.1222MusgroveElizabeth A.2JonesMarc D.12NourseCraig23JamiesonNigel B.222GrahamJanet S.222BiankinAndrew V.1222ChangDavid K.1222JamiesonNigel B.222GrahamJanet S.222The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 370 Victoria Street, Darlinghurst, Sydney, New South Wales, AustraliaWolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United KingdomQueensland Centre for Medical Genomics, Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, AustraliaRoyal North Shore Hospital, St Leonards, New South Wales, AustraliaBankstown Hospital, Bankstown, New South Wales, AustraliaLiverpool Hospital, Liverpool, New South Wales, AustraliaWestmead Hospital, Westmead, New South Wales, AustraliaRoyal Prince Alfred Hospital, Camperdown, New South Wales, AustraliaFremantle Hospital, Fremantle, Western Australia, AustraliaSir Charles Gairdner Hospital, Nedlands, Western Australia, AustraliaSt John of God Healthcare, Subiaco, Western Australia, AustraliaRoyal Adelaide Hospital, Adelaide, South Australia, AustraliaFlinders Medical Centre, Bedford Park, South Australia, AustraliaGreenslopes Private Hospital, Greenslopes, Queensland, AustraliaEnvoi Pathology, Herston, Queensland, AustraliaPrincess Alexandria Hospital, Woolloongabba, Queensland, AustraliaAustin Hospital, Heidelberg, Victoria, AustraliaVictorian Cancer Biobank, Carlton, Victoria, AustraliaJohns Hopkins Medical Institute, Baltimore, MarylandARC-NET Center for Applied Research on Cancer, University of Verona, Verona, ItalyUniversity of California, San Francisco, San Francisco, CaliforniaGreater Glasgow and Clyde National Health Service, Glasgow, United Kingdom
- Australian Pancreas Genome, Darlinghurst, Australia
| | - Amber Johns
- The Kinghorn Cancer Centre, Darlinghurst and Garvan Institute of Medical Research, Sydney, Australia
| | - Marina Pajic
- The Kinghorn Cancer Centre, Darlinghurst and Garvan Institute of Medical Research, Sydney, Australia
| | - Fieke E.M. Froeling
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York,Epigenetics Unit, Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, London, United Kingdom
| | - Phillip Beer
- Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Elizabeth A. Musgrove
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | | | - Alan Ashworth
- Department of Pathology, Queen Elizabeth University Hospital, Glasgow, United Kingdom,University of California–San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Margaret C. Frame
- Medical Research Council Institute of Genetics and Molecular Medicine, Edinburgh Cancer Research UK Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Howard C. Crawford
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Diane M. Simeone
- Pancreatic Cancer Center, Perlmutter Cancer Center, New York University Langone Health, New York, New York
| | - Chris Lord
- Cancer Research UK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, Florida
| | | | - David A. Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Susanna L. Cooke
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Nigel B. Jamieson
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom,West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Jennifer P. Morton
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom,Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Owen J. Sansom
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom,Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
| | - Peter J. Bailey
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Andrew V. Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom,West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, United Kingdom,South Western Sydney Clinical School, Faculty of Medicine, University of New South Wales, Liverpool, Australia,Andrew V. Biankin, MD, PhD, Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow, Scotland G61 1BD, United Kingdom fax: +44 141 330 5834.
| | - David K. Chang
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom,West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, United Kingdom,South Western Sydney Clinical School, Faculty of Medicine, University of New South Wales, Liverpool, Australia,Correspondence Address correspondence to: David K. Chang, MD, PhD, Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow, Scotland G61 1BD, United Kingdom fax: +44 141 330 5834.
| |
Collapse
|
13
|
DNA damage response and breast cancer development: Possible therapeutic applications of ATR, ATM, PARP, BRCA1 inhibition. DNA Repair (Amst) 2020; 98:103032. [PMID: 33494010 DOI: 10.1016/j.dnarep.2020.103032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
Breast cancer is the most common and significant cancers in females regarding the loss of life quality. Similar to other cancers, one of the etiologic factors in breast cancer is DNA damage. A plethora of molecules are responsible for sensing DNA damage and mediating actions which lead to DNA repair, senescence, cell cycle arrest and if damage is unbearable to apoptosis. In each of these, aberrations leading to unrepaired damage was resulted in uncontrolled proliferation and cancer. Another cellular function is autophagy defined as a process eliminating of unnecessary proteins in stress cases involved in pathogenesis of cancer. Knowing their role in cancer, scholars have tried to develop strategies in order to target DDR and autophagy. Further, the interactions of DDR and autophagy plus their regulatory role on each other have been focused simultaneously. The present review study has aimed to illustrate the importance of DDR and autophagy in breast cancer according to the related studies and uncover the relation between DDR and autophagy and its significance in breast cancer therapy.
Collapse
|
14
|
Qiu Z, Fa P, Liu T, Prasad CB, Ma S, Hong Z, Chan ER, Wang H, Li Z, He K, Wang QE, Williams TM, Yan C, Sizemore ST, Narla G, Zhang J. A Genome-Wide Pooled shRNA Screen Identifies PPP2R2A as a Predictive Biomarker for the Response to ATR and CHK1 Inhibitors. Cancer Res 2020; 80:3305-3318. [PMID: 32522823 PMCID: PMC7518641 DOI: 10.1158/0008-5472.can-20-0057] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/17/2020] [Accepted: 06/04/2020] [Indexed: 01/18/2023]
Abstract
There is currently a lack of precise predictive biomarkers for patient selection in clinical trials of inhibitors targeting replication stress (RS) response proteins ATR and CHK1. The objective of this study was to identify novel predictive biomarkers for the response to these agents in treating non-small cell lung cancer (NSCLC). A genome-wide loss-of-function screen revealed that tumor suppressor PPP2R2A, a B regulatory subunit of protein phosphatase 2 (PP2A), determines sensitivity to CHK1 inhibition. A synthetic lethal interaction between PPP2R2A deficiency and ATR or CHK1 inhibition was observed in NSCLC in vitro and in vivo and was independent of p53 status. ATR and CHK1 inhibition resulted in significantly increased levels of RS and altered replication dynamics, particularly in PPP2R2A-deficient NSCLC cells. Mechanistically, PPP2R2A negatively regulated translation of oncogene c-Myc protein. c-Myc activity was required for PPP2R2A deficiency-induced alterations of replication initiation/RS and sensitivity to ATR/CHK1 inhibitors. We conclude that PPP2R2A deficiency elevates RS by upregulating c-Myc activity, rendering cells reliant on the ATR/CHK1 axis for survival. Our studies show a novel synthetic lethal interaction and identify PPP2R2A as a potential new predictive biomarker for patient stratification in the clinical use of ATR and CHK1 inhibitors. SIGNIFICANCE: This study reveals new approaches to specifically target PPP2R2A-deficient lung cancer cells and provides a novel biomarker that will significantly improve treatment outcome with ATR and CHK1 inhibitors.
Collapse
MESH Headings
- Animals
- Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors
- Biomarkers, Tumor/deficiency
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Non-Small-Cell Lung/chemistry
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Cell Line, Tumor
- Checkpoint Kinase 1/antagonists & inhibitors
- DNA Damage
- DNA Replication
- Drug Resistance, Neoplasm
- Female
- Gene Knockdown Techniques
- Genes, p53
- Genome-Wide Association Study
- Heterografts
- Humans
- Lung Neoplasms/chemistry
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Male
- Mice
- Mice, Nude
- Protein Phosphatase 2/deficiency
- Protein Phosphatase 2/genetics
- Protein Phosphatase 2/metabolism
- Proto-Oncogene Proteins c-myc/metabolism
- RNA, Small Interfering
Collapse
Affiliation(s)
- Zhaojun Qiu
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Pengyan Fa
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Tao Liu
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Chandra B Prasad
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Shanhuai Ma
- University of Rochester, Rochester, New York
| | - Zhipeng Hong
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Ernest R Chan
- Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Zaibo Li
- Department of Pathology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Kai He
- Department of Internal Medicine, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Qi-En Wang
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Terence M Williams
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Chunhong Yan
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Steven T Sizemore
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio
| | - Goutham Narla
- Department of Medicine, University of Michigan, Ann Arbor, Michigan
| | - Junran Zhang
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio.
| |
Collapse
|
15
|
Lai J, Yang H, Zhu Y, Ruan M, Huang Y, Zhang Q. MiR-7-5p-mediated downregulation of PARP1 impacts DNA homologous recombination repair and resistance to doxorubicin in small cell lung cancer. BMC Cancer 2019; 19:602. [PMID: 31215481 PMCID: PMC6582543 DOI: 10.1186/s12885-019-5798-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/05/2019] [Indexed: 12/13/2022] Open
Abstract
Background Chemo-resistance is one of the major challenges in the therapy of small cell lung cancer (SCLC). Multiple mechanisms are thought to be involved in chemo-resistance during SCLC treatment, but unfortunately, these mechanisms have not been well elucidated. Herein, we investigated the role of miRNA in the resistance of SCLC cells to doxorubicin (Dox). Methods MiRNA microarray analysis revealed that several miRNAs, including miR-7-5p, were specifically decreased in Dox-resistant SCLC cells (H69AR) compared to parental cells (H69). The expression level of miR-7-5p was confirmed by qRT-PCR in Dox-resistant cells (H69AR and H446AR cells) and their parental cells. Bioinformatic analysis indicated that poly ADP-ribose polymerase 1 (PARP1) is a direct target of miR-7-5p. The binding sites of miR-7-5p in the PARP1 3′ UTR were verified by luciferase reporter and Western blot assays. To investigate the role of miR-7-5p in the chemo-resistance of SCLC cells to doxorubicin, mimic or inhibitor of miR-7-5p was transfected into SCLC cells, and the effect of miR-7-5p on homologous recombination (HR) repair was analyzed by HR reporter assays. Furthermore, the expression of HR repair factors (Rad51 and BRCA1) induced by doxorubicin was detected by Western blot and immunofluorescent staining in H446AR cells transfected with miR-7-5p mimic. Results The expression level of miR-7-5p was remarkably reduced (4-fold) in Dox-resistant SCLC cells (H69AR and H446AR cells) compared with that in parental cells (H69 and H446 cells). Poly ADP-ribose polymerase 1 (PARP1) is a direct target of miR-7-5p, and PARP1 expression was downregulated by miR-7-5p. MiR-7-5p impeded Dox-induced HR repair by inhibiting the expression of HR repair factors (Rad51 and BRCA1) that resulted in resensitizing SCLC cells to doxorubicin. Conclusions Our findings provide evidence that miR-7-5p targets PARP1 to exert its suppressive effects on HR repair, indicating that the alteration of the expression of miR-7-5p may be a promising strategy for overcoming chemo-resistance in SCLC therapy. Electronic supplementary material The online version of this article (10.1186/s12885-019-5798-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jinzhi Lai
- Department of Oncology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China.,Institute of Immunotherapy, Fujian Medical University, Fuzhou, 350108, Fujian, China
| | - Hainan Yang
- Department of Ultrasound, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Yanyang Zhu
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, 350108, Fujian, China
| | - Mei Ruan
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, 350108, Fujian, China
| | - Yayu Huang
- Department of Oncology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Qiuyu Zhang
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, 350108, Fujian, China.
| |
Collapse
|
16
|
Savva C, De Souza K, Ali R, Rakha EA, Green AR, Madhusudan S. Clinicopathological significance of ataxia telangiectasia-mutated (ATM) kinase and ataxia telangiectasia-mutated and Rad3-related (ATR) kinase in MYC overexpressed breast cancers. Breast Cancer Res Treat 2019; 175:105-115. [PMID: 30746633 PMCID: PMC6491658 DOI: 10.1007/s10549-018-05113-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 12/18/2018] [Indexed: 12/20/2022]
Abstract
PURPOSE MYC transcription factor has critical roles in cell growth, proliferation, metabolism, differentiation, transformation and angiogenesis. MYC overexpression is seen in about 15% of breast cancers and linked to aggressive phenotypes. MYC overexpression also induces oxidative stress and replication stress in cells. ATM signalling and ATR-mediated signalling are critical for MYC-induced DNA damage response. Whether ATM and ATR expressions influence clinical outcomes in MYC overexpressed breast cancers is unknown. METHODS We investigated ATM, ATR and MYC at the transcriptional level [Molecular Taxonomy of Breast Cancer International Consortium cohort (n = 1950)] and at the protein level in the Nottingham series comprising 1650 breast tumours. We correlated ATM, ATR and MYC expressions to clinicopathological features and survival outcomes. RESULTS In MYC over expressed tumours, high ATR or low ATM levels were associated with aggressive breast cancer features such as higher tumour grade, de-differentiation, pleomorphism, high mitotic index, high-risk Nottingham Prognostic Index, triple negative and basal-like breast cancers (all adjusted p values < 0.05). Tumours with low ATM or high ATR levels in conjunction with MYC overexpression also have worse overall breast cancer-specific survival (BCSS) (p value < 0.05). CONCLUSIONS We conclude that ATR/ATM-directed stratification and personalisation of therapy may be feasible in MYC overexpressed breast cancer.
Collapse
Affiliation(s)
- Constantinos Savva
- Department of Oncology, Nottingham University Hospitals, Nottingham, NG5 1PB, UK
| | - Karen De Souza
- Department of Oncology, Nottingham University Hospitals, Nottingham, NG5 1PB, UK
| | - Reem Ali
- Translational Oncology, Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, Academic Unit of Oncology, School of Medicine, University of Nottingham, Nottingham, NG51 PB, UK
| | - Emad A Rakha
- Department of Pathology, Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, NG5 1PB, UK
| | - Andrew R Green
- Department of Pathology, Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, NG5 1PB, UK.
| | - Srinivasan Madhusudan
- Department of Oncology, Nottingham University Hospitals, Nottingham, NG5 1PB, UK.
- Translational Oncology, Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, Academic Unit of Oncology, School of Medicine, University of Nottingham, Nottingham, NG51 PB, UK.
| |
Collapse
|
17
|
Post AEM, Bussink J, Sweep FCGJ, Span PN. Changes in DNA Damage Repair Gene Expression and Cell Cycle Gene Expression Do Not Explain Radioresistance in Tamoxifen-Resistant Breast Cancer. Oncol Res 2019; 28:33-40. [PMID: 31046897 PMCID: PMC7851527 DOI: 10.3727/096504019x15555794826018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Tamoxifen-induced radioresistance, reported in vitro, might pose a problem for patients who receive neoadjuvant tamoxifen treatment and subsequently receive radiotherapy after surgery. Previous studies suggested that DNA damage repair or cell cycle genes are involved, and could therefore be targeted to preclude the occurrence of cross-resistance. We aimed to characterize the observed cross-resistance by investigating gene expression of DNA damage repair genes and cell cycle genes in estrogen receptor-positive MCF-7 breast cancer cells that were cultured to tamoxifen resistance. RNA sequencing was performed, and expression of genes characteristic for several DNA damage repair pathways was investigated, as well as expression of genes involved in different phases of the cell cycle. The association of differentially expressed genes with outcome after radiotherapy was assessed in silico in a large breast cancer cohort. None of the DNA damage repair pathways showed differential gene expression in tamoxifen-resistant cells compared to wild-type cells. Two DNA damage repair genes were more than two times upregulated (NEIL1 and EME2), and three DNA damage repair genes were more than two times downregulated (PCNA, BRIP1, and BARD1). However, these were not associated with outcome after radiotherapy in the TCGA breast cancer cohort. Genes involved in G1, G1/S, G2, and G2/M phases were lower expressed in tamoxifen-resistant cells compared to wild-type cells. Individual genes that were more than two times upregulated (MAPK13) or downregulated (E2F2, CKS2, GINS2, PCNA, MCM5, and EIF5A2) were not associated with response to radiotherapy in the patient cohort investigated. We assessed the expression of DNA damage repair genes and cell cycle genes in tamoxifen-resistant breast cancer cells. Though several genes in both pathways were differentially expressed, these could not explain the cross-resistance for irradiation in these cells, since no association to response to radiotherapy in the TCGA breast cancer cohort was found.
Collapse
Affiliation(s)
- Annemarie E M Post
- Radboud University Medical Center, Department of Radiation Oncology, Radiotherapy and OncoImmunology LaboratoryNijmegenThe Netherlands
| | - Johan Bussink
- Radboud University Medical Center, Department of Radiation Oncology, Radiotherapy and OncoImmunology LaboratoryNijmegenThe Netherlands
| | - Fred C G J Sweep
- Radboud University Medical Center, Department of Laboratory MedicineNijmegenThe Netherlands
| | - Paul N Span
- Radboud University Medical Center, Department of Radiation Oncology, Radiotherapy and OncoImmunology LaboratoryNijmegenThe Netherlands
| |
Collapse
|
18
|
Yadav P, Shankar BS. Radio resistance in breast cancer cells is mediated through TGF-β signalling, hybrid epithelial-mesenchymal phenotype and cancer stem cells. Biomed Pharmacother 2019; 111:119-130. [DOI: 10.1016/j.biopha.2018.12.055] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 12/11/2018] [Accepted: 12/14/2018] [Indexed: 12/20/2022] Open
|
19
|
Miao W, Fan M, Huang M, Li JJ, Wang Y. Targeted Profiling of Heat Shock Proteome in Radioresistant Breast Cancer Cells. Chem Res Toxicol 2019; 32:326-332. [PMID: 30596229 DOI: 10.1021/acs.chemrestox.8b00330] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Breast cancer is the most commonly diagnosed cancer and the second leading cause of cancer death in women. Radioresistance remains one of the most critical barriers in radiation therapy for breast cancer. In this study, we employed a parallel-reaction monitoring (PRM)-based targeted proteomic method to examine the reprogramming of the heat shock proteome during the development of radioresistance in breast cancer. In particular, we investigated the differential expression of heat shock proteins (HSPs) in two pairs of matched parental/radioresistant breast cancer cell lines. We were able to quantify 43 and 42 HSPs in the MCF-7 and MDA-MB-231 pairs of cell lines, respectively. By analyzing the commonly altered proteins, we found that several members of the HSP70 and HSP40 subfamilies of HSPs exhibited substantially altered expression upon development of radioresistance. Moreover, the expression of HSPB8 is markedly elevated in the radioresistant lines relative to the parental MCF-7 and MDA-MB-231 cells. Together, our PRM-based targeted proteomics method revealed the reprogramming of the heat shock proteome during the development of radioresistance in breast cancer cells and offered potential targets for sensitizing breast cancer cells toward radiation therapy.
Collapse
|
20
|
Wu M, Pang JS, Sun Q, Huang Y, Hou JY, Chen G, Zeng JJ, Feng ZB. The clinical significance of CHEK1 in breast cancer: a high-throughput data analysis and immunohistochemical study. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:1-20. [PMID: 31933717 PMCID: PMC6944032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 11/26/2018] [Indexed: 06/10/2023]
Abstract
Breast cancer (BC) is a kind of malignant cancer that seriously threatens women's health. Research scientists have found that BC occurs as the result of multiple effects of the external environment and internal genetic changes. Cell cycle checkpoint kinase 1 (CHEK1) is a crucial speed limit point in the cell cycle. Alterations of CHEK1 have been found in various tumors but are rarely reported or verified in BC. By mining database information, a large amount of mRNA and protein data was collected and meta-analyzed. Also, in-house immunohistochemistry was carried out to validate the results of the CHEK1 expression levels. Relative clinical features of BC patients were calculated with the CHEK1 expression levels to determine their diagnostic value. The mRNA levels of CHEK1 were higher in 1,089 cases of BC tissues than in 291 cases of non-BC tissues. We observed that the mRNA levels of CHEK1 are related to the clinical stages of BC patients (P = 0.008) and are also significant for overall survival (HR = 1.6, P = 0.0081). Using the immunohistochemistry method, we calculated and confirmed, using Fisher's exact test (P < 0.001), that a high-level CHEK1 protein is exhibited in BC tissues. Overexpressed CHEK1 mRNA promotes the occurrence of BC. Also, up-regulated CHEK1 could serve as an independent risk biomarker in BC patients' prognoses.
Collapse
Affiliation(s)
- Mei Wu
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, Guangxi Zhuang Autonomous Region, P. R. China
| | - Jin-Shu Pang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, Guangxi Zhuang Autonomous Region, P. R. China
| | - Qi Sun
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, Guangxi Zhuang Autonomous Region, P. R. China
| | - Yu Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi University of Traditional Chinese MedicineNanning 530021, Guangxi Zhuang Autonomous Region, P. R. China
| | - Jia-Yin Hou
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, Guangxi Zhuang Autonomous Region, P. R. China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, Guangxi Zhuang Autonomous Region, P. R. China
| | - Jing-Jing Zeng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, Guangxi Zhuang Autonomous Region, P. R. China
| | - Zhen-Bo Feng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, Guangxi Zhuang Autonomous Region, P. R. China
| |
Collapse
|
21
|
Lee WH, Chen LC, Lee CJ, Huang CC, Ho YS, Yang PS, Ho CT, Chang HL, Lin IH, Chang HW, Liu YR, Wu CH, Tu SH. DNA primase polypeptide 1 (PRIM1) involves in estrogen-induced breast cancer formation through activation of the G2/M cell cycle checkpoint. Int J Cancer 2018; 144:615-630. [PMID: 30097999 DOI: 10.1002/ijc.31788] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 07/24/2018] [Indexed: 12/22/2022]
Abstract
The DNA primase polypeptide 1 (PRIM1) is responsible for synthesizing small RNA primers for Okazaki fragments generated during discontinuous DNA replication. PRIM1 mRNA expression levels in breast tumor samples were detected by real-time PCR analysis. Xenografted tumor model was established to study the carcinogenic role of PRIM1 and its potential therapeutic applications. The average PRIM1 mRNA (copy number × 103 /μg) expression was 4.7-fold higher in tumors than in normal tissue (*p = 0.005, n = 254). PRIM1 was detected preferentially at a higher level (>40-fold) in poorly differentiated tumor tissues (n = 46) compared with more highly differentiated tumors tissues (n = 10) (*p = 0.005). Poor overall survival rate was correlated to the estrogen receptor positive (ER+, n = 20) patients with higher PRIM1 expression when compare to the ER- (n = 10) patients (Chi Square test, p = 0.03). Stable expression of PRIM1-siRNA in the ER+ BT-474 cells-xenograft tumors significantly reduced tumor volume in SCID mice (*p = 0.005). The anti-tumoral effects of inotilone isolated from Phellinus linteus was tested and had significant effects on the inhibition of PRIM1 protein expression in ER+ breast cancer cells. In vivo study was performed by administering inotilone (10 mg/kg, twice a week for 6 weeks), which resulted in significantly reduced BT-474-xenografted tumor growth volume compared with control (n =5 per group, *p < 0.05). This study provides evidences for the prognostic effects of PRIM1 with poor overall survival rate in the ER+ patients and will be valuable to test for therapeutic purpose.
Collapse
Affiliation(s)
- Wei-Hwa Lee
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pathology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Li-Ching Chen
- Breast Medical Center, Taipei Medical University Hospital, Taipei, Taiwan.,Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Jung Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chi-Cheng Huang
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan.,Department of Surgery, Fu-Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Yuan-Soon Ho
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Laboratory Medicine, Taipei Medical University Hospital, Taipei, Taiwan.,School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Po-Sheng Yang
- Department of Surgery, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ, USA
| | - Hang-Lung Chang
- Department of General Surgery, En Chu Kong Hospital, New Taipei City, Taiwan
| | - I-Hsuan Lin
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hui-Wen Chang
- Department of Laboratory Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yun-Ru Liu
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.,Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Chih-Hsiung Wu
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of General Surgery, En Chu Kong Hospital, New Taipei City, Taiwan
| | - Shih-Hsin Tu
- Breast Medical Center, Taipei Medical University Hospital, Taipei, Taiwan.,Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
22
|
Yang X, Pan Y, Qiu Z, Du Z, Zhang Y, Fa P, Gorityala S, Ma S, Li S, Chen C, Wang H, Xu Y, Yan C, Ruth K, Ma Z, Zhang J. RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast Cancer and CHEK1 Inhibitor Efficacy in Breast Cancer Cells. Clin Cancer Res 2018; 24:1629-1643. [PMID: 29326282 DOI: 10.1158/1078-0432.ccr-17-2242] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/10/2017] [Accepted: 01/05/2018] [Indexed: 01/20/2023]
Abstract
Purpose: (i) To investigate the expression of the E3 ligase, RNF126, in human invasive breast cancer and its links with breast cancer outcomes; and (ii) to test the hypothesis that RNF126 determines the efficacy of inhibitors targeting the cell-cycle checkpoint kinase, CHEK1.Experimental Design: A retrospective analysis by immunohistochemistry (IHC) compared RNF126 staining in 110 invasive breast cancer and 78 paired adjacent normal tissues with clinicopathologic data. Whether RNF126 controls CHEK1 expression was determined by chromatin immunoprecipitation and a CHEK1 promoter driven luciferase reporter. Staining for these two proteins by IHC using tissue microarrays was also conducted. Cell killing/replication stress induced by CHEK1 inhibition was evaluated in cells, with or without RNF126 knockdown, by MTT/colony formation, replication stress biomarker immunostaining and DNA fiber assays.Results: RNF126 protein expression was elevated in breast cancer tissue samples. RNF126 was associated with a poor clinical outcome after multivariate analysis and was an independent predictor. RNF126 promotes CHEK1 transcript expression. Critically, a strong correlation between RNF126 and CHEK1 proteins was identified in breast cancer tissue and cell lines. The inhibition of CHEK1 induced a greater cell killing and a higher level of replication stress in breast cancer cells expressing RNF126 compared to RNF126 depleted cells.Conclusions: RNF126 protein is highly expressed in invasive breast cancer tissue. The high expression of RNF126 is an independent predictor of a poor prognosis in invasive breast cancer and is considered a potential biomarker of a cancer's responsiveness to CHEK1 inhibitors. CHEK1 inhibition targets breast cancer cells expressing higher levels of RNF126 by enhancing replication stress. Clin Cancer Res; 24(7); 1629-43. ©2018 AACR.
Collapse
Affiliation(s)
- Xiaosong Yang
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University, Cleveland, Ohio.,Department of Breast Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - You Pan
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University, Cleveland, Ohio.,Department of Breast Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhaojun Qiu
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Zhanwen Du
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Yao Zhang
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Pengyan Fa
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | | | - Shanhuai Ma
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Shunqiang Li
- Division of Oncology Breast Oncology Section, Washington University Medical School, St. Louis, Missouri
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Yan Xu
- Department of Chemistry, Cleveland State University, Cleveland, Ohio.,Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Chunhong Yan
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Keri Ruth
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio.,Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Zhefu Ma
- Department of Breast Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China. .,Department of Breast Surgery & Plastic Surgery, Cancer Hospital of China Medical University, Shenyang, China
| | - Junran Zhang
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University, Cleveland, Ohio. .,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
23
|
Yao S, Fan LYN, Lam EWF. The FOXO3-FOXM1 axis: A key cancer drug target and a modulator of cancer drug resistance. Semin Cancer Biol 2017; 50:77-89. [PMID: 29180117 PMCID: PMC6565931 DOI: 10.1016/j.semcancer.2017.11.018] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/30/2017] [Accepted: 11/23/2017] [Indexed: 12/11/2022]
Abstract
The FOXO3 and FOXM1 forkhead box transcription factors, functioning downstream of the essential PI3K-Akt, Ras-ERK and JNK/p38MAPK signalling cascades, are crucial for cell proliferation, differentiation, cell survival, senescence, DNA damage repair and cell cycle control. The development of resistance to both conventional and newly emerged molecularly targeted therapies is a major challenge confronting current cancer treatment in the clinic. Intriguingly, the mechanisms of resistance to ‘classical’ cytotoxic chemotherapeutics and to molecularly targeted therapies are invariably linked to deregulated signalling through the FOXO3 and FOXM1 transcription factors. This is owing to the involvement of FOXO3 and FOXM1 in the regulation of genes linked to crucial drug action-related cellular processes, including stem cell renewal, DNA repair, cell survival, drug efflux, and deregulated mitosis. A better understanding of the mechanisms regulating the FOXO3-FOXM1 axis, as well as their downstream transcriptional targets and functions, may render these proteins reliable and early diagnostic/prognostic factors as well as crucial therapeutic targets for cancer treatment and importantly, for overcoming chemotherapeutic drug resistance.
Collapse
Affiliation(s)
- Shang Yao
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Lavender Yuen-Nam Fan
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Eric Wing-Fai Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK.
| |
Collapse
|
24
|
Qiu Z, Oleinick NL, Zhang J. ATR/CHK1 inhibitors and cancer therapy. Radiother Oncol 2017; 126:450-464. [PMID: 29054375 DOI: 10.1016/j.radonc.2017.09.043] [Citation(s) in RCA: 226] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 08/01/2017] [Accepted: 09/30/2017] [Indexed: 02/06/2023]
Abstract
The cell cycle checkpoint proteins ataxia-telangiectasia-mutated-and-Rad3-related kinase (ATR) and its major downstream effector checkpoint kinase 1 (CHK1) prevent the entry of cells with damaged or incompletely replicated DNA into mitosis when the cells are challenged by DNA damaging agents, such as radiation therapy (RT) or chemotherapeutic drugs, that are the major modalities to treat cancer. This regulation is particularly evident in cells with a defective G1 checkpoint, a common feature of cancer cells, due to p53 mutations. In addition, ATR and/or CHK1 suppress replication stress (RS) by inhibiting excess origin firing, particularly in cells with activated oncogenes. Those functions of ATR/CHK1 make them ideal therapeutic targets. ATR/CHK1 inhibitors have been developed and are currently used either as single agents or paired with radiotherapy or a variety of genotoxic chemotherapies in preclinical and clinical studies. Here, we review the status of the development of ATR and CHK1 inhibitors. We also discuss the potential mechanisms by which ATR and CHK1 inhibition induces cell killing in the presence or absence of exogenous DNA damaging agents, such as RT and chemotherapeutic agents. Lastly, we discuss synthetic lethality interactions between the inhibition of ATR/CHK1 and defects in other DNA damage response (DDR) pathways/genes.
Collapse
Affiliation(s)
- Zhaojun Qiu
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University, Cleveland, USA
| | - Nancy L Oleinick
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University, Cleveland, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, USA
| | - Junran Zhang
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University, Cleveland, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, USA.
| |
Collapse
|
25
|
Emptage RP, Schoenberger MJ, Ferguson KM, Marmorstein R. Intramolecular autoinhibition of checkpoint kinase 1 is mediated by conserved basic motifs of the C-terminal kinase-associated 1 domain. J Biol Chem 2017; 292:19024-19033. [PMID: 28972186 DOI: 10.1074/jbc.m117.811265] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/21/2017] [Indexed: 11/06/2022] Open
Abstract
Precise control of the cell cycle allows for timely repair of genetic material prior to replication. One factor intimately involved in this process is checkpoint kinase 1 (Chk1), a DNA damage repair inducing Ser/Thr protein kinase that contains an N-terminal kinase domain and a C-terminal regulatory region consisting of a ∼100-residue linker followed by a putative kinase-associated 1 (KA1) domain. We report the crystal structure of the human Chk1 KA1 domain, demonstrating striking structural homology with other sequentially diverse KA1 domains. Separately purified Chk1 kinase and KA1 domains are intimately associated in solution, which results in inhibition of Chk1 kinase activity. Using truncation mutants and site-directed mutagenesis, we define the inhibitory face of the KA1 domain as a series of basic residues residing on two conserved regions of the primary structure. These findings point to KA1-mediated intramolecular autoinhibition as a key regulatory mechanism of human Chk1, and provide new therapeutic possibilities with which to attack this validated oncology target with small molecules.
Collapse
Affiliation(s)
- Ryan P Emptage
- From the Department of Biochemistry and Biophysics and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104,
| | - Megan J Schoenberger
- the Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, and
| | - Kathryn M Ferguson
- the Department of Pharmacology and Cancer Biology Institute, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Ronen Marmorstein
- From the Department of Biochemistry and Biophysics and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, .,the Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, and
| |
Collapse
|
26
|
King AR, Corso CD, Chen EM, Song E, Bongiorni P, Chen Z, Sundaram RK, Bindra RS, Saltzman WM. Local DNA Repair Inhibition for Sustained Radiosensitization of High-Grade Gliomas. Mol Cancer Ther 2017; 16:1456-1469. [PMID: 28566437 DOI: 10.1158/1535-7163.mct-16-0788] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 04/14/2017] [Accepted: 05/16/2017] [Indexed: 11/16/2022]
Abstract
High-grade gliomas, such as glioblastoma (GBM) and diffuse intrinsic pontine glioma (DIPG), are characterized by an aggressive phenotype with nearly universal local disease progression despite multimodal treatment, which typically includes chemotherapy, radiotherapy, and possibly surgery. Radiosensitizers that have improved the effects of radiotherapy for extracranial tumors have been ineffective for the treatment of GBM and DIPG, in part due to poor blood-brain barrier penetration and rapid intracranial clearance of small molecules. Here, we demonstrate that nanoparticles can provide sustained drug release and minimal toxicity. When administered locally, these nanoparticles conferred radiosensitization in vitro and improved survival in rats with intracranial gliomas when delivered concurrently with a 5-day course of fractionated radiotherapy. Compared with previous work using locally delivered radiosensitizers and cranial radiation, our approach, based on the rational selection of agents and a clinically relevant radiation dosing schedule, produces the strongest synergistic effects between chemo- and radiotherapy approaches to the treatment of high-grade gliomas. Mol Cancer Ther; 16(8); 1456-69. ©2017 AACR.
Collapse
Affiliation(s)
- Amanda R King
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Christopher D Corso
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Evan M Chen
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Eric Song
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Paul Bongiorni
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Zhe Chen
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Ranjini K Sundaram
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Ranjit S Bindra
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut. .,Department of Experimental Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut.
| |
Collapse
|
27
|
Targeting the ATR-CHK1 Axis in Cancer Therapy. Cancers (Basel) 2017; 9:cancers9050041. [PMID: 28448462 PMCID: PMC5447951 DOI: 10.3390/cancers9050041] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/23/2017] [Accepted: 04/25/2017] [Indexed: 12/14/2022] Open
Abstract
Targeting the DNA damage response (DDR) is a new therapeutic approach in cancer that shows great promise for tumour selectivity. Key components of the DDR are the ataxia telangiectasia mutated and Rad3 related (ATR) and checkpoint kinase 1 (CHK1) kinases. This review article describes the role of ATR and its major downstream target, CHK1, in the DDR and why cancer cells are particularly reliant on the ATR-CHK1 pathway, providing the rationale for targeting these kinases, and validation of this hypothesis by genetic manipulation. The recent development of specific inhibitors and preclinical data using these inhibitors not only as chemosensitisers and radiosensitisers but also as single agents to exploit specific pathologies of tumour cells is described. These potent and specific inhibitors have now entered clinical trial and early results are presented.
Collapse
|