1
|
Athwal H, Kochiyanil A, Bhat V, Allan AL, Parsyan A. Centrosomes and associated proteins in pathogenesis and treatment of breast cancer. Front Oncol 2024; 14:1370565. [PMID: 38606093 PMCID: PMC11007099 DOI: 10.3389/fonc.2024.1370565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 03/04/2024] [Indexed: 04/13/2024] Open
Abstract
Breast cancer is the most prevalent malignancy among women worldwide. Despite significant advances in treatment, it remains one of the leading causes of female mortality. The inability to effectively treat advanced and/or treatment-resistant breast cancer demonstrates the need to develop novel treatment strategies and targeted therapies. Centrosomes and their associated proteins have been shown to play key roles in the pathogenesis of breast cancer and thus represent promising targets for drug and biomarker development. Centrosomes are fundamental cellular structures in the mammalian cell that are responsible for error-free execution of cell division. Centrosome amplification and aberrant expression of its associated proteins such as Polo-like kinases (PLKs), Aurora kinases (AURKs) and Cyclin-dependent kinases (CDKs) have been observed in various cancers, including breast cancer. These aberrations in breast cancer are thought to cause improper chromosomal segregation during mitosis, leading to chromosomal instability and uncontrolled cell division, allowing cancer cells to acquire new genetic changes that result in evasion of cell death and the promotion of tumor formation. Various chemical compounds developed against PLKs and AURKs have shown meaningful antitumorigenic effects in breast cancer cells in vitro and in vivo. The mechanism of action of these inhibitors is likely related to exacerbation of numerical genomic instability, such as aneuploidy or polyploidy. Furthermore, growing evidence demonstrates enhanced antitumorigenic effects when inhibitors specific to centrosome-associated proteins are used in combination with either radiation or chemotherapy drugs in breast cancer. This review focuses on the current knowledge regarding the roles of centrosome and centrosome-associated proteins in breast cancer pathogenesis and their utility as novel targets for breast cancer treatment.
Collapse
Affiliation(s)
- Harjot Athwal
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Arpitha Kochiyanil
- Faculty of Science, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Vasudeva Bhat
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
| | - Alison L. Allan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Armen Parsyan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Division of General Surgery, Department of Surgery, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Surgery, St. Joseph’s Health Care London and London Health Sciences Centre, London, ON, Canada
| |
Collapse
|
2
|
Chao Y, Chen Y, Zheng W, Demanelis K, Liu Y, Connelly JA, Wang H, Li S, Wang QJ. Synthetic lethal combination of CHK1 and WEE1 inhibition for treatment of castration-resistant prostate cancer. Oncogene 2024; 43:789-803. [PMID: 38273024 DOI: 10.1038/s41388-024-02939-z] [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] [Received: 11/05/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/27/2024]
Abstract
WEE1 and CHEK1 (CHK1) kinases are critical regulators of the G2/M cell cycle checkpoint and DNA damage response pathways. The WEE1 inhibitor AZD1775 and the CHK1 inhibitor SRA737 are in clinical trials for various cancers, but have not been thoroughly examined in prostate cancer, particularly castration-resistant (CRPC) and neuroendocrine prostate cancers (NEPC). Our data demonstrated elevated WEE1 and CHK1 expressions in CRPC and NEPC cell lines and patient samples. AZD1775 resulted in rapid and potent cell killing with comparable IC50s across different prostate cancer cell lines, while SRA737 displayed time-dependent progressive cell killing with 10- to 20-fold differences in IC50s. Notably, their combination synergistically reduced the viability of all CRPC cell lines and tumor spheroids in a concentration- and time-dependent manner. Importantly, in a transgenic mouse model of NEPC, both agents alone or in combination suppressed tumor growth, improved overall survival, and reduced the incidence of distant metastases, with SRA737 exhibiting remarkable single agent anticancer activity. Mechanistically, SRA737 synergized with AZD1775 by blocking AZD1775-induced feedback activation of CHK1 in prostate cancer cells, resulting in increased mitotic entry and accumulation of DNA damage. In summary, this preclinical study shows that CHK1 inhibitor SRA737 alone and its combination with AZD1775 offer potential effective treatments for CRPC and NEPC.
Collapse
Affiliation(s)
- Yapeng Chao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yuzhou Chen
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Wenxiao Zheng
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kathryn Demanelis
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA, USA
| | - Yu Liu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jaclyn A Connelly
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hong Wang
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Qiming Jane Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Zhou M, Duan L, Chen J, Li Y, Yin Z, Song S, Cao Y, Luo P, Hu F, Yang G, Xu J, Liao T, Jin Y. The dynamic role of nucleoprotein SHCBP1 in the cancer cell cycle and its potential as a synergistic target for DNA-damaging agents in cancer therapy. Cell Commun Signal 2024; 22:131. [PMID: 38365687 PMCID: PMC10874017 DOI: 10.1186/s12964-024-01513-0] [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: 09/25/2023] [Accepted: 02/01/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Malignant tumours seriously threaten human life and health, and effective treatments for cancer are still being explored. The ability of SHC SH2 domain-binding protein 1 (SHCBP1) to induce cell cycle disturbance and inhibit tumour growth has been increasingly studied, but its dynamic role in the tumour cell cycle and corresponding effects leading to mitotic catastrophe and DNA damage have rarely been studied. RESULTS In this paper, we found that the nucleoprotein SHCBP1 exhibits dynamic spatiotemporal expression during the tumour cell cycle, and SHCBP1 knockdown slowed cell cycle progression by inducing spindle disorder, as reflected by premature mitotic entry and multipolar spindle formation. This dysfunction was caused by G2/M checkpoint impairment mediated by downregulated WEE1 kinase and NEK7 (a member of the mammalian NIMA-related kinase family) expression and upregulated centromere/kinetochore protein Zeste White 10 (ZW10) expression. Moreover, both in vivo and in vitro experiments confirmed the significant inhibitory effects of SHCBP1 knockdown on tumour growth. Based on these findings, SHCBP1 knockdown in combination with low-dose DNA-damaging agents had synergistic tumouricidal effects on tumour cells. In response to this treatment, tumour cells were forced into the mitotic phase with considerable unrepaired DNA lesions, inducing mitotic catastrophe. These synergistic effects were attributed not only to the abrogation of the G2/M checkpoint and disrupted spindle function but also to the impairment of the DNA damage repair system, as demonstrated by mass spectrometry-based proteomic and western blotting analyses. Consistently, patients with low SHCBP1 expression in tumour tissue were more sensitive to radiotherapy. However, SHCBP1 knockdown combined with tubulin-toxic drugs weakened the killing effect of the drugs on tumour cells, which may guide the choice of chemotherapeutic agents in clinical practice. CONCLUSION In summary, we elucidated the role of the nucleoprotein SHCBP1 in tumour cell cycle progression and described a novel mechanism by which SHCBP1 regulates tumour progression and through which targeting SHCBP1 increases sensitivity to DNA-damaging agent therapy, indicating its potential as a cancer treatment.
Collapse
Affiliation(s)
- Mei Zhou
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Engineering Research Center for Tumour-Targeted Biochemotherapy, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Limin Duan
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Engineering Research Center for Tumour-Targeted Biochemotherapy, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiangbin Chen
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Engineering Research Center for Tumour-Targeted Biochemotherapy, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Yumei Li
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Engineering Research Center for Tumour-Targeted Biochemotherapy, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Zhengrong Yin
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Engineering Research Center for Tumour-Targeted Biochemotherapy, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Siwei Song
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Engineering Research Center for Tumour-Targeted Biochemotherapy, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Yaqi Cao
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Engineering Research Center for Tumour-Targeted Biochemotherapy, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Ping Luo
- Department of Translational Medicine Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Fan Hu
- Medical Subcenter of HUST Analytical & Testing Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Guanghai Yang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Juanjuan Xu
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Engineering Research Center for Tumour-Targeted Biochemotherapy, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Tingting Liao
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Engineering Research Center for Tumour-Targeted Biochemotherapy, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
- Hubei Province Engineering Research Center for Tumour-Targeted Biochemotherapy, Union HospitalTongji Medical CollegeHuazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| |
Collapse
|
4
|
Abd El-Hafeez T, Shams MY, Elshaier YAMM, Farghaly HM, Hassanien AE. Harnessing machine learning to find synergistic combinations for FDA-approved cancer drugs. Sci Rep 2024; 14:2428. [PMID: 38287066 PMCID: PMC10825182 DOI: 10.1038/s41598-024-52814-w] [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: 11/09/2023] [Accepted: 01/24/2024] [Indexed: 01/31/2024] Open
Abstract
Combination therapy is a fundamental strategy in cancer chemotherapy. It involves administering two or more anti-cancer agents to increase efficacy and overcome multidrug resistance compared to monotherapy. However, drug combinations can exhibit synergy, additivity, or antagonism. This study presents a machine learning framework to classify and predict cancer drug combinations. The framework utilizes several key steps including data collection and annotation from the O'Neil drug interaction dataset, data preprocessing, stratified splitting into training and test sets, construction and evaluation of classification models to categorize combinations as synergistic, additive, or antagonistic, application of regression models to predict combination sensitivity scores for enhanced predictions compared to prior work, and the last step is examination of drug features and mechanisms of action to understand synergy behaviors for optimal combinations. The models identified combination pairs most likely to synergize against different cancers. Kinase inhibitors combined with mTOR inhibitors, DNA damage-inducing drugs or HDAC inhibitors showed benefit, particularly for ovarian, melanoma, prostate, lung and colorectal carcinomas. Analysis highlighted Gemcitabine, MK-8776 and AZD1775 as frequently synergizing across cancer types. This machine learning framework provides a valuable approach to uncover more effective multi-drug regimens.
Collapse
Affiliation(s)
- Tarek Abd El-Hafeez
- Department of Computer Science, Faculty of Science, Minia University, El-Minia, Egypt.
- Computer Science Unit, Deraya University, El-Minia, Egypt.
| | - Mahmoud Y Shams
- Faculty of Artificial Intelligence, Kafrelsheikh University, Kafr El-Sheikh, Egypt
- Scientific Research Group in Egypt (SRGE), Cairo, Egypt
| | - Yaseen A M M Elshaier
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, Menoufia, Egypt
| | - Heba Mamdouh Farghaly
- Department of Computer Science, Faculty of Science, Minia University, El-Minia, Egypt
| | - Aboul Ella Hassanien
- Faculty of Computers and Artificial Intelligence, Cairo University, Cairo, Egypt.
- Scientific Research Group in Egypt (SRGE), Cairo, Egypt.
| |
Collapse
|
5
|
Zhang C, Peng K, Liu Q, Huang Q, Liu T. Adavosertib and beyond: Biomarkers, drug combination and toxicity of WEE1 inhibitors. Crit Rev Oncol Hematol 2024; 193:104233. [PMID: 38103761 DOI: 10.1016/j.critrevonc.2023.104233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023] Open
Abstract
WEE1 kinase is renowned as an S-G2 checkpoint inhibitor activated by ATR-CHK1 in response to replication stress. WEE1 inhibition enhances replication stress and effectively circumvents checkpoints into mitosis, which triggers significant genetic impairs and culminates in cell death. This approach has been validated clinically for its promising anti-tumor efficacy across various cancer types, notably in cases of ovarian cancers. Nonetheless, the initial stage of clinical trials has shown that the first-in-human WEE1 inhibitor adavosertib is limited by dose-limiting adverse events. As a result, recent efforts have been made to explore predictive biomarkers and smart combination schedules to alleviate adverse effects. In this review, we focused on the exploration of therapeutic biomarkers, as well as schedules of combination utilizing WEE1 inhibitors and canonical anticancer drugs, according to the latest preclinical and clinical studies, indicating that the optimal application of WEE1 inhibitors will likely be as part of dose-reducing combination and be tailored to specific patient populations.
Collapse
Affiliation(s)
- Chi Zhang
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ke Peng
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Liu
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qihong Huang
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Tianshu Liu
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
O'Brien S, Ubhi T, Wolf L, Gandhi K, Lin S, Chaudary N, Dhani NC, Milosevic M, Brown GW, Angers S. FBXW7-loss Sensitizes Cells to ATR Inhibition Through Induced Mitotic Catastrophe. CANCER RESEARCH COMMUNICATIONS 2023; 3:2596-2607. [PMID: 38032106 PMCID: PMC10734389 DOI: 10.1158/2767-9764.crc-23-0306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/27/2023] [Accepted: 11/28/2023] [Indexed: 12/01/2023]
Abstract
FBXW7 is a commonly mutated tumor suppressor gene that functions to regulate numerous oncogenes involved in cell-cycle regulation. Genome-wide CRISPR fitness screens identified a signature of DNA repair and DNA damage response genes as required for the growth of FBXW7-knockout cells. Guided by these findings, we show that FBXW7-mutant cells have high levels of replication stress, which results in a genotype-specific vulnerability to inhibition of the ATR signaling pathway, as these mutant cells become heavily reliant on a robust S-G2 checkpoint. ATR inhibition induces an accelerated S-phase, leading to mitotic catastrophe and cell death caused by the high replication stress present in FBXW7-/- cells. In addition, we provide evidence in cell and organoid studies, and mining of publicly available high-throughput drug screening efforts, that this genotype-specific vulnerability extends to multiple types of cancer, providing a rational means of identifying responsive patients for targeted therapy. SIGNIFICANCE We have elucidated the synthetic lethal interactions between FBXW7 mutation and DNA damage response genes, and highlighted the potential of ATR inhibitors as targeted therapies for cancers harboring FBXW7 alterations.
Collapse
Affiliation(s)
- Siobhan O'Brien
- Department of Biochemistry, University of Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada
| | - Tajinder Ubhi
- Department of Biochemistry, University of Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada
| | - Lucie Wolf
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada
| | - Krishna Gandhi
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada
| | - Sichun Lin
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada
| | - Naz Chaudary
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Ontario Cancer Institute, Toronto, Ontario, Canada
| | | | - Michael Milosevic
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, University of Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Ontario, Canada
| | - Grant W. Brown
- Department of Biochemistry, University of Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada
| | - Stephane Angers
- Department of Biochemistry, University of Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada
| |
Collapse
|
7
|
Wang Q, Chao Y, Chen Y, Zheng W, Demanelis K, Liu Y, Connelly J, Wang H. Synthetic lethal combination of CHK1 and WEE1 inhibition for treatment of castration-resistant prostate cancer. RESEARCH SQUARE 2023:rs.3.rs-3564450. [PMID: 37987002 PMCID: PMC10659531 DOI: 10.21203/rs.3.rs-3564450/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
WEE1 and CHEK1 (CHK1) kinases are critical regulators of the G2/M cell cycle checkpoint and DNA damage response pathways. The WEE1 inhibitor AZD1775 and the CHK1 inhibitor SRA737 are in clinical trials for various cancers, but have not been examined in prostate cancer, particularly castration-resistant (CRPC) and neuroendocrine prostate cancers (NEPC). Our data demonstrated elevated WEE1 and CHK1 expressions in CRPC/NEPC cell lines and patient samples. AZD1775 resulted in rapid and potent cell killing with comparable IC50s across different prostate cancer cell lines, while SRA737 displayed time-dependent progressive cell killing with 10- to 20-fold differences in IC50s. Notably, their combination synergistically reduced the viability of all CRPC cell lines and tumor spheroids in a concentration- and time-dependent manner. Importantly, in a transgenic mouse model of NEPC, both agents alone or in combination suppressed tumor growth, improved overall survival, and reduced the incidence of distant metastases, with SRA737 exhibiting remarkable single agent anticancer activity. Mechanistically, SRA737 synergized with AZD1775 by blocking AZD1775-induced feedback activation of CHK1 in prostate cancer cells, resulting in increased mitotic entry and accumulation of DNA damage. In summary, this preclinical study shows that CHK1 inhibitor SRA737 alone and its combination with AZD1775 offer potential effective treatments for CRPC and NEPC.
Collapse
Affiliation(s)
| | | | | | | | | | - Yu Liu
- University of Pittsburgh Cancer Institute
| | | | - Hong Wang
- University of Pittsburgh Cancer Institute
| |
Collapse
|
8
|
Nair NU, Greninger P, Zhang X, Friedman AA, Amzallag A, Cortez E, Sahu AD, Lee JS, Dastur A, Egan RK, Murchie E, Ceribelli M, Crowther GS, Beck E, McClanaghan J, Klump-Thomas C, Boisvert JL, Damon LJ, Wilson KM, Ho J, Tam A, McKnight C, Michael S, Itkin Z, Garnett MJ, Engelman JA, Haber DA, Thomas CJ, Ruppin E, Benes CH. A landscape of response to drug combinations in non-small cell lung cancer. Nat Commun 2023; 14:3830. [PMID: 37380628 PMCID: PMC10307832 DOI: 10.1038/s41467-023-39528-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/14/2023] [Indexed: 06/30/2023] Open
Abstract
Combination of anti-cancer drugs is broadly seen as way to overcome the often-limited efficacy of single agents. The design and testing of combinations are however very challenging. Here we present a uniquely large dataset screening over 5000 targeted agent combinations across 81 non-small cell lung cancer cell lines. Our analysis reveals a profound heterogeneity of response across the tumor models. Notably, combinations very rarely result in a strong gain in efficacy over the range of response observable with single agents. Importantly, gain of activity over single agents is more often seen when co-targeting functionally proximal genes, offering a strategy for designing more efficient combinations. Because combinatorial effect is strongly context specific, tumor specificity should be achievable. The resource provided, together with an additional validation screen sheds light on major challenges and opportunities in building efficacious combinations against cancer and provides an opportunity for training computational models for synergy prediction.
Collapse
Affiliation(s)
- Nishanth Ulhas Nair
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Xiaohu Zhang
- Howard Hughes Medical Institute, Bethesda, MD, USA
| | - Adam A Friedman
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Arnaud Amzallag
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eliane Cortez
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Avinash Das Sahu
- University of New Mexico, Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Joo Sang Lee
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Anahita Dastur
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Regina K Egan
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ellen Murchie
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Erin Beck
- Howard Hughes Medical Institute, Bethesda, MD, USA
| | | | | | | | - Leah J Damon
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Jeffrey Ho
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Angela Tam
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Sam Michael
- Howard Hughes Medical Institute, Bethesda, MD, USA
| | - Zina Itkin
- Howard Hughes Medical Institute, Bethesda, MD, USA
| | - Mathew J Garnett
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, CB10 1SA, UK
| | | | - Daniel A Haber
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Bethesda, MD, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institute of Health, Rockville, MD, 20850, USA
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Cyril H Benes
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
9
|
Xu X, Kumari R, Zhou J, Chen J, Mao B, Wang J, Zheng M, Tu X, An X, Chen X, Zhang L, Tian X, Wang H, Dong X, Bao Z, Guo S, Ouyang X, Shang L, Wang F, Yan X, Zhang R, Vries RGJ, Clevers H, Li QX. A living biobank of matched pairs of patient-derived xenografts and organoids for cancer pharmacology. PLoS One 2023; 18:e0279821. [PMID: 36602988 PMCID: PMC9815646 DOI: 10.1371/journal.pone.0279821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023] Open
Abstract
Patient-derived tumor xenograft (PDX)/organoid (PDO), driven by cancer stem cells (CSC), are considered the most predictive models for translational oncology. Large PDX collections reflective of patient populations have been created and used extensively to test various investigational therapies, including population-trials as surrogate subjects in vivo. PDOs are recognized as in vitro surrogates for patients amenable for high-throughput screening (HTS). We have built a biobank of carcinoma PDX-derived organoids (PDXOs) by converting an existing PDX library and confirmed high degree of similarities between PDXOs and parental PDXs in genomics, histopathology and pharmacology, suggesting "biological equivalence or interchangeability" between the two. Here we demonstrate the applications of PDXO biobank for HTS "matrix" screening for both lead compounds and indications, immune cell co-cultures for immune-therapies and engineering enables in vitro/in vivo imaging. This large biobank of >550 matched pairs of PDXs/PDXOs across different cancers could become powerful tools for the future cancer drug discovery.
Collapse
Affiliation(s)
- Xiaoxi Xu
- Crown Bioscience Inc., Beijing, China
| | - Rajendra Kumari
- Crown Bioscience Inc., San Diego, California, United States of America
| | - Jun Zhou
- Crown Bioscience Inc., Taicang City, Jiangsu, China
| | - Jing Chen
- Crown Bioscience Inc., San Diego, California, United States of America
| | - Binchen Mao
- Crown Bioscience Inc., Taicang City, Jiangsu, China
| | | | | | - Xiaolong Tu
- Crown Bioscience Inc., Taicang City, Jiangsu, China
| | - Xiaoyu An
- Crown Bioscience Inc., San Diego, California, United States of America
| | | | | | - Xiaoli Tian
- Shanghai Yihao Biological Technology, Xuhui District, Shanghai, China
| | - Haojie Wang
- Suzhou NeoLogics Bioscience Co, LTD, Suzhou, China
| | - Xin Dong
- Suzhou NeoLogics Bioscience Co, LTD, Suzhou, China
| | | | - Sheng Guo
- Crown Bioscience Inc., Taicang City, Jiangsu, China
| | | | | | - Fei Wang
- Crown Bioscience Inc., Taicang City, Jiangsu, China
| | - Xuefei Yan
- Crown Bioscience Inc., Taicang City, Jiangsu, China
| | - Rui Zhang
- Crown Bioscience Inc., Taicang City, Jiangsu, China
| | - Robert G. J. Vries
- Hubrecht Organoid Technology (HUB), Utrecht, The Netherlands
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Utrecht, The Netherlands
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Utrecht, The Netherlands
| | - Qi-Xiang Li
- Crown Bioscience Inc., San Diego, California, United States of America
| |
Collapse
|
10
|
Targeting the DNA damage response: PARP inhibitors and new perspectives in the landscape of cancer treatment. Crit Rev Oncol Hematol 2021; 168:103539. [PMID: 34800653 DOI: 10.1016/j.critrevonc.2021.103539] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/26/2021] [Accepted: 11/15/2021] [Indexed: 12/27/2022] Open
Abstract
Cancer derives from alterations of pathways responsible for cell survival, differentiation and proliferation. Dysfunctions of mechanisms protecting genome integrity can promote oncogenesis but can also be exploited as therapeutic target. Poly-ADP-Ribose-Polymerase (PARP)-inhibitors, the first approved targeted agents able to tackle DNA damage response (DDR), have demonstrated antitumor activity, particularly when homologous recombination impairment is present. Despite the relevant results achieved, a large proportion of patients fail to obtain durable responses. The development of innovative treatments, able to overcome resistance and ensure long-lasting benefit for a wider population is still an unmet need. Moreover, improvement in biomarker assays is necessary to properly identify patients who can benefit from DDR targeting agents. Here we summarize the main DDR pathways, explain the current role of PARP inhibitors in cancer therapy and illustrate new therapeutic strategies targeting the DDR, focusing on the combinations of PARP inhibitors with other agents and on cell-cycle checkpoint inhibitors.
Collapse
|
11
|
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
|
12
|
van Bijsterveldt L, Durley SC, Maughan TS, Humphrey TC. The Challenge of Combining Chemo- and Radiotherapy with Checkpoint Kinase Inhibitors. Clin Cancer Res 2021; 27:937-962. [PMID: 33257428 DOI: 10.1158/1078-0432.ccr-20-3358] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/10/2020] [Accepted: 11/20/2020] [Indexed: 11/16/2022]
Abstract
Preclinical models of cancer have demonstrated enhanced efficacy of cell-cycle checkpoint kinase inhibitors when used in combination with genotoxic agents. This combination therapy is predicted to be exquisitely toxic to cells with a deficient G1-S checkpoint or cells with a genetic predisposition leading to intrinsic DNA replication stress, as these cancer cells become fully dependent on the intra-S and G2-M checkpoints for DNA repair and cellular survival. Therefore, abolishing remaining cell-cycle checkpoints after damage leads to increased cell death in a tumor cell-specific fashion. However, the preclinical success of these drug combinations is not consistently replicated in clinical trials. Here, we provide a perspective on the translation of preclinical studies into rationally designed clinical studies. We will discuss successes and failures of current treatment combinations and drug regimens and provide a detailed overview of all clinical trials using ATR, CHK1, or WEE1 inhibitors in combination with genotoxic agents. This highlights the need for revised patient stratification and the use of appropriate pharmacodynamic biomarkers to improve the success rate of clinical trials.
Collapse
Affiliation(s)
- Linda van Bijsterveldt
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Samuel C Durley
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Tim S Maughan
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Timothy C Humphrey
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
13
|
Pharmacological Inhibition of WEE1 Potentiates the Antitumoral Effect of the dl922-947 Oncolytic Virus in Malignant Mesothelioma Cell Lines. Int J Mol Sci 2020; 21:ijms21197333. [PMID: 33020398 PMCID: PMC7582744 DOI: 10.3390/ijms21197333] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/24/2022] Open
Abstract
Malignant mesothelioma (MM) is a very aggressive asbestos-related cancer, for which no therapy proves to be effective. We have recently shown that the oncolytic adenovirus dl922-947 had antitumor effects in MM cell lines and murine xenografts. Previous studies demonstrated that dl922-947-induced host cell cycle checkpoint deregulation and consequent DNA lesions associated with the virus efficacy. However, the cellular DNA damage response (DDR) can counteract this virus action. Therefore, we assessed whether AZD1775, an inhibitor of the G2/M DNA damage checkpoint kinase WEE1, could enhance MM cell sensitivity to dl922-947. Through cell viability assays, we found that AZD1775 synergized with dl922-947 selectively in MM cell lines and increased dl922-947-induced cell death, which showed hallmarks of apoptosis (annexinV-positivity, caspase-dependency, BCL-XL decrease, chromatin condensation). Predictably, dl922-947 and/or AZD1775 activated the DDR, as indicated by increased levels of three main DDR players: phosphorylated histone H2AX (γ-H2AX), phospho-replication protein A (RPA)32, phospho-checkpoint kinase 1 (CHK1). Dl922-947 also increased inactive Tyr-15-phosphorylated cyclin-dependent kinase 1 (CDK1), a key WEE1 substrate, which is indicative of G2/M checkpoint activation. This increase in phospho-CDK1 was effectively suppressed by AZD1775, thus suggesting that this compound could, indeed, abrogate the dl922-947-induced DNA damage checkpoint in MM cells. Overall, our data suggest that the dl922-947-AZD1775 combination could be a feasible strategy against MM.
Collapse
|
14
|
Neizer-Ashun F, Bhattacharya R. Reality CHEK: Understanding the biology and clinical potential of CHK1. Cancer Lett 2020; 497:202-211. [PMID: 32991949 DOI: 10.1016/j.canlet.2020.09.016] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/26/2020] [Accepted: 09/20/2020] [Indexed: 12/13/2022]
Abstract
The DNA damage response enables cells to cope with various stresses that threaten genomic integrity. A critical component of this response is the serine/threonine kinase CHK1 which is encoded by the CHEK1 gene. Originally identified as a regulator of the G2/M checkpoint, CHK1 has since been shown to play important roles in DNA replication, mitotic progression, DNA repair, and overall cell cycle regulation. However, the potential of CHK1 as a cancer therapy has not been realized clinically. Herein we expound our current understanding of the principal roles of CHK1 and highlight different avenues for CHK1 targeting in cancer therapy.
Collapse
Affiliation(s)
- Fiifi Neizer-Ashun
- Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, 73104, United States
| | - Resham Bhattacharya
- Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, 73104, United States; Department of Obstetrics and Gynecology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, United States; Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK, 73104, United States.
| |
Collapse
|
15
|
Simonenko V, Lu X, Roesch E, Mutisya D, Shao C, Sun Q, Patterson-Orazem A, McNair M, Shanmuganathan A, Lu P, Evans DM. A novel siRNA-gemcitabine construct as a potential therapeutic for treatment of pancreatic cancer. NAR Cancer 2020; 2:zcaa016. [PMID: 34316688 PMCID: PMC8209983 DOI: 10.1093/narcan/zcaa016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/02/2020] [Accepted: 07/28/2020] [Indexed: 12/17/2022] Open
Abstract
The non-nucleoside analog gemcitabine has been the standard of care for treating pancreatic cancer. The drug shows good potency in pancreatic cancer cells in vitro but, due to poor bioavailability, requires administration in large doses by infusion and this systemic exposure results in significant toxicity for the patient. Genes have been identified that, when silenced by siRNA, synergize with gemcitabine treatment and offer a means of reducing the gemcitabine dosage required for efficacy. However, benefiting from the synergism between the two agents requires that the gemcitabine and siRNA penetrate the same cells. To ensure co-delivery, we incorporated gemcitabine covalently within siRNAs against targets synergistic with gemcitabine (CHK1 or RAD17). We demonstrated that specific bases within an siRNA can be replaced with gemcitabine to increase efficacy. The result is a single drug molecule that simultaneously co-delivers gemcitabine and a synergistic siRNA. The siRNA–gemcitabine constructs demonstrate a 5–30-fold improvement in potency compared with gemcitabine alone. Co-delivering a CHK1 siRNA–gemcitabine construct together with a WEE1 siRNA resulted in a 10-fold improvement in IC50 compared with gemcitabine alone. These constructs demonstrate efficacy across a wide array of pancreatic tumor cells and may represent a novel therapeutic approach for treating pancreatic cancer.
Collapse
Affiliation(s)
- Vera Simonenko
- Sirnaomics Inc., Suite 280, 401 Professional Drive, Gaithersburg, MD 20879, USA
| | - Xiaoyong Lu
- Sirnaomics Inc., Suite 280, 401 Professional Drive, Gaithersburg, MD 20879, USA
| | - Eric Roesch
- Sirnaomics Inc., Suite 280, 401 Professional Drive, Gaithersburg, MD 20879, USA
| | - Daniel Mutisya
- Sirnaomics Inc., Suite 280, 401 Professional Drive, Gaithersburg, MD 20879, USA
| | - Chunbo Shao
- Sirnaomics Inc., Suite 280, 401 Professional Drive, Gaithersburg, MD 20879, USA
| | - Qian Sun
- Sirnaomics Inc., Suite 280, 401 Professional Drive, Gaithersburg, MD 20879, USA
| | | | - Marcus McNair
- Sirnaomics Inc., Suite 280, 401 Professional Drive, Gaithersburg, MD 20879, USA
| | | | - Patrick Lu
- Sirnaomics Inc., Suite 280, 401 Professional Drive, Gaithersburg, MD 20879, USA
| | - David M Evans
- Sirnaomics Inc., Suite 280, 401 Professional Drive, Gaithersburg, MD 20879, USA
| |
Collapse
|
16
|
Ghelli Luserna Di Rorà A, Bocconcelli M, Ferrari A, Terragna C, Bruno S, Imbrogno E, Beeharry N, Robustelli V, Ghetti M, Napolitano R, Chirumbolo G, Marconi G, Papayannidis C, Paolini S, Sartor C, Simonetti G, Yen TJ, Martinelli G. Synergism Through WEE1 and CHK1 Inhibition in Acute Lymphoblastic Leukemia. Cancers (Basel) 2019; 11:cancers11111654. [PMID: 31717700 PMCID: PMC6895917 DOI: 10.3390/cancers11111654] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/16/2019] [Accepted: 10/23/2019] [Indexed: 12/30/2022] Open
Abstract
Introduction: Screening for synthetic lethality markers has demonstrated that the inhibition of the cell cycle checkpoint kinases WEE1 together with CHK1 drastically affects stability of the cell cycle and induces cell death in rapidly proliferating cells. Exploiting this finding for a possible therapeutic approach has showed efficacy in various solid and hematologic tumors, though not specifically tested in acute lymphoblastic leukemia. Methods: The efficacy of the combination between WEE1 and CHK1 inhibitors in B and T cell precursor acute lymphoblastic leukemia (B/T-ALL) was evaluated in vitro and ex vivo studies. The efficacy of the therapeutic strategy was tested in terms of cytotoxicity, induction of apoptosis, and changes in cell cycle profile and protein expression using B/T-ALL cell lines. In addition, the efficacy of the drug combination was studied in primary B-ALL blasts using clonogenic assays. Results: This study reports, for the first time, the efficacy of the concomitant inhibition of CHK1/CHK2 and WEE1 in ALL cell lines and primary leukemic B-ALL cells using two selective inhibitors: PF-0047736 (CHK1/CHK2 inhibitor) and AZD-1775 (WEE1 inhibitor). We showed strong synergism in the reduction of cell viability, proliferation and induction of apoptosis. The efficacy of the combination was related to the induction of early S-phase arrest and to the induction of DNA damage, ultimately triggering cell death. We reported evidence that the efficacy of the combination treatment is independent from the activation of the p53-p21 pathway. Moreover, gene expression analysis on B-ALL primary samples showed that Chek1 and Wee1 are significantly co-expressed in samples at diagnosis (Pearson r = 0.5770, p = 0.0001) and relapse (Pearson r= 0.8919; p = 0.0001). Finally, the efficacy of the combination was confirmed by the reduction in clonogenic survival of primary leukemic B-ALL cells. Conclusion: Our findings suggest that the combination of CHK1 and WEE1 inhibitors may be a promising therapeutic strategy to be tested in clinical trials for adult ALL.
Collapse
Affiliation(s)
| | - Matteo Bocconcelli
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, 40138 Bologna, Italy
| | - Anna Ferrari
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (A.G.L.D.R.)
| | - Carolina Terragna
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, 40138 Bologna, Italy
| | - Samantha Bruno
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, 40138 Bologna, Italy
| | - Enrica Imbrogno
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (A.G.L.D.R.)
| | | | - Valentina Robustelli
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, 40138 Bologna, Italy
| | - Martina Ghetti
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (A.G.L.D.R.)
| | - Roberta Napolitano
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (A.G.L.D.R.)
| | - Gabriella Chirumbolo
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, 40138 Bologna, Italy
| | - Giovanni Marconi
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, 40138 Bologna, Italy
| | - Cristina Papayannidis
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, 40138 Bologna, Italy
| | - Stefania Paolini
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, 40138 Bologna, Italy
| | - Chiara Sartor
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, 40138 Bologna, Italy
| | - Giorgia Simonetti
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (A.G.L.D.R.)
- Correspondence:
| | - Timothy J. Yen
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111-2497, USA
| | - Giovanni Martinelli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (A.G.L.D.R.)
| |
Collapse
|
17
|
Byrum AK, Vindigni A, Mosammaparast N. Defining and Modulating 'BRCAness'. Trends Cell Biol 2019; 29:740-751. [PMID: 31362850 DOI: 10.1016/j.tcb.2019.06.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/24/2019] [Accepted: 06/27/2019] [Indexed: 02/08/2023]
Abstract
The concept of 'BRCAness' defines the pathogenesis and vulnerability of multiple cancers. The canonical definition of BRCAness is a defect in homologous recombination repair, mimicking BRCA1 or BRCA2 loss. In turn, BRCA-deficient cells utilize error-prone DNA-repair pathways, causing increased genomic instability, which may be responsible for their sensitivity to DNA damaging agents and poly-(ADP)-ribose polymerase inhibitors (PARPis). However, recent work has expanded the mechanistic basis of BRCAness, to include defects in replication fork protection (RFP). Here, we broaden the definition of BRCAness to include RFP and regulatory mechanisms that cause synthetic lethality with PARPis. We highlight these recent discoveries, which include mechanisms of RFP regulation, DNA damage checkpoint proteins, as well as kinases that regulate BRCA1/2 function. Importantly, many of these emerging mechanisms may be targeted for inhibition with small molecule inhibitors, thus inducing BRCAness in a much larger subset of BRCA-proficient tumors, with significant translational potential.
Collapse
Affiliation(s)
- Andrea K Byrum
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Alessandro Vindigni
- Division of Oncology, Department of Internal Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA.
| | - Nima Mosammaparast
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
18
|
Xu Z, Ni R, Chen Y. Targeting breast cancer stem cells by a self-assembled, aptamer-conjugated DNA nanotrain with preloading doxorubicin. Int J Nanomedicine 2019; 14:6831-6842. [PMID: 31695364 PMCID: PMC6717853 DOI: 10.2147/ijn.s200482] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 05/15/2019] [Indexed: 12/30/2022] Open
Abstract
Background Cancer relapse and metastasis is an obstacle to the treatment of breast cancer. Breast cancer stem cells (BCSCs), which can evade the killing effect of traditional chemotherapies, such as doxorubicin (DOX), may contribute to cancer development. Therefore, it is necessary to develop novel drugs that can target and eliminate BCSCs. While multiple strategies have been conceived, they are normally limited by the low drug loading capacity. Purpose An aptamer-conjugated DNA nanotrain TA6NT-AKTin-DOX, which consists of a CD44 aptamer TA6, DNA building blocks M1 and M2 conjugated with an AKT inhibitor peptide AKTin individually and DOX, was designed. Methods This DNA nanotrain was prepared through hybridization chain reactionand this highly ordered DNA duplex has plenty of sites where DOX and AKTin can be intercalated or anchored. By performing on MCF-7 BCSCs and tumors by xenografting BCSCs into nude mice, efficacy of the newly prepared drug was evaluated and compared with that of free DOX and various DNA nanotrains. Results TA6NT-AKTin-DOX showed better efficacy both in vitro and in vivo. To some extent, the enhanced efficacy could be attributed to the targeting effect of TA6 and the high drug loading capacity of the nanotrain (~20 DOX molecules). Besides, a synergistic response was demonstrated by combining DOX with AKTin, probably due to that the anchored AKTin can reverse the drug resistance of BCSCs including apoptosis resistance and ABC transporters overexpression via the AKT signaling pathway. Conclusion The aptamer-conjugated DNA nanotrain TA6NT-AKTin-DOX demonstrated its targeting capability to BCSCs.
Collapse
Affiliation(s)
- Zhiyuan Xu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, People's Republic of China
| | - Ronghua Ni
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, People's Republic of China
| | - Yun Chen
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, People's Republic of China.,State Key Laboratory of Reproductive Medicine, Nanjing 210029, People's Republic of China
| |
Collapse
|
19
|
Erber J, Steiner JD, Isensee J, Lobbes LA, Toschka A, Beleggia F, Schmitt A, Kaiser RWJ, Siedek F, Persigehl T, Hucho T, Reinhardt HC. Dual Inhibition of GLUT1 and the ATR/CHK1 Kinase Axis Displays Synergistic Cytotoxicity in KRAS-Mutant Cancer Cells. Cancer Res 2019; 79:4855-4868. [PMID: 31405847 DOI: 10.1158/0008-5472.can-18-3959] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 06/18/2019] [Accepted: 08/06/2019] [Indexed: 11/16/2022]
Abstract
The advent of molecularly targeted therapeutic agents has opened a new era in cancer therapy. However, many tumors rely on nondruggable cancer-driving lesions. In addition, long-lasting clinical benefits from single-agent therapies rarely occur, as most of the tumors acquire resistance over time. The identification of targeted combination regimens interfering with signaling through oncogenically rewired pathways provides a promising approach to enhance efficacy of single-agent-targeted treatments. Moreover, combination drug therapies might overcome the emergence of drug resistance. Here, we performed a focused flow cytometry-based drug synergy screen and identified a novel synergistic interaction between GLUT1-mediated glucose transport and the cell-cycle checkpoint kinases ATR and CHK1. Combined inhibition of CHK1/GLUT1 or ATR/GLUT1 robustly induced apoptosis, particularly in RAS-mutant cancer cells. Mechanistically, combined inhibition of ATR/CHK1 and GLUT1 arrested sensitive cells in S-phase and led to the accumulation of genotoxic damage, particularly in S-phase. In vivo, simultaneous inhibition of ATR and GLUT1 significantly reduced tumor volume gain in an autochthonous mouse model of KrasG12D -driven soft tissue sarcoma. Taken together, these findings pave the way for combined inhibition of GLUT1 and ATR/CHK1 as a therapeutic approach for KRAS-driven cancers. SIGNIFICANCE: Dual targeting of the DNA damage response and glucose transport synergistically induces apoptosis in KRAS-mutant cancer, suggesting this combination treatment for clinical validation in KRAS-stratified tumor patients.
Collapse
Affiliation(s)
- Johanna Erber
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Dusseldorf, Center for Molecular Medicine Cologne, CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
| | - Joachim D Steiner
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Dusseldorf, Center for Molecular Medicine Cologne, CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jörg Isensee
- Translational Pain Research, Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Leonard A Lobbes
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Dusseldorf, Center for Molecular Medicine Cologne, CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - André Toschka
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Dusseldorf, Center for Molecular Medicine Cologne, CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Filippo Beleggia
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Dusseldorf, Center for Molecular Medicine Cologne, CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Anna Schmitt
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Dusseldorf, Center for Molecular Medicine Cologne, CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Rainer W J Kaiser
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Florian Siedek
- Institute of Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Thorsten Persigehl
- Institute of Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Tim Hucho
- Translational Pain Research, Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Hans C Reinhardt
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Dusseldorf, Center for Molecular Medicine Cologne, CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
20
|
Margue C, Philippidou D, Kozar I, Cesi G, Felten P, Kulms D, Letellier E, Haan C, Kreis S. Kinase inhibitor library screening identifies synergistic drug combinations effective in sensitive and resistant melanoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:56. [PMID: 30728057 PMCID: PMC6364417 DOI: 10.1186/s13046-019-1038-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 01/13/2019] [Indexed: 12/19/2022]
Abstract
Background Melanoma is the most aggressive and deadly form of skin cancer with increasing case numbers worldwide. The development of inhibitors targeting mutated BRAF (found in around 60% of melanoma patients) has markedly improved overall survival of patients with late-stage tumors, even more so when combined with MEK inhibitors targeting the same signaling pathway. However, invariably patients become resistant to this targeted therapy resulting in rapid progression with treatment-refractory disease. The purpose of this study was the identification of new kinase inhibitors that do not lead to the development of resistance in combination with BRAF inhibitors (BRAFi), or that could be of clinical benefit as a 2nd line treatment for late-stage melanoma patients that have already developed resistance. Methods We have screened a 274-compound kinase inhibitor library in 3 BRAF mutant melanoma cell lines (each one sensitive or made resistant to 2 distinct BRAFi). The screening results were validated by dose-response studies and confirmed the killing efficacies of many kinase inhibitors. Two different tools were applied to investigate and quantify potential synergistic effects of drug combinations: the Chou-Talalay method and the Synergyfinder application. In order to exclude that resistance to the new treatments might occur at later time points, synergistic combinations were administered to fluorescently labelled parental and resistant cells over a period of > 10 weeks. Results Eight inhibitors targeting Wee1, Checkpoint kinase 1/2, Aurora kinase, MEK, Polo-like kinase, PI3K and Focal adhesion kinase killed melanoma cells synergistically when combined with a BRAFi. Additionally, combination of a Wee1 and Chk inhibitor showed synergistic killing effects not only on sensitive cell lines, but also on intrinsically BRAFi- and treatment induced-resistant melanoma cells. First in vivo studies confirmed these observations. Interestingly, continuous treatment with several of these drugs, alone or in combination, did not lead to emergence of resistance. Conclusions Here, we have identified new, previously unexplored (in the framework of BRAFi resistance) inhibitors that have an effect not only on sensitive but also on BRAFi-resistant cells. These promising combinations together with the new immunotherapies could be an important step towards improved 1st and 2nd line treatments for late-stage melanoma patients. Electronic supplementary material The online version of this article (10.1186/s13046-019-1038-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christiane Margue
- Life Sciences Research Unit, University of Luxembourg, 6, av. du Swing, L-4367, Belvaux, Luxembourg
| | - Demetra Philippidou
- Life Sciences Research Unit, University of Luxembourg, 6, av. du Swing, L-4367, Belvaux, Luxembourg
| | - Ines Kozar
- Life Sciences Research Unit, University of Luxembourg, 6, av. du Swing, L-4367, Belvaux, Luxembourg
| | - Giulia Cesi
- Life Sciences Research Unit, University of Luxembourg, 6, av. du Swing, L-4367, Belvaux, Luxembourg
| | - Paul Felten
- Life Sciences Research Unit, University of Luxembourg, 6, av. du Swing, L-4367, Belvaux, Luxembourg
| | - Dagmar Kulms
- Experimental Dermatology, Department of Dermatology, Technical University Dresden, Dresden, Germany
| | - Elisabeth Letellier
- Life Sciences Research Unit, University of Luxembourg, 6, av. du Swing, L-4367, Belvaux, Luxembourg
| | - Claude Haan
- Life Sciences Research Unit, University of Luxembourg, 6, av. du Swing, L-4367, Belvaux, Luxembourg
| | - Stephanie Kreis
- Life Sciences Research Unit, University of Luxembourg, 6, av. du Swing, L-4367, Belvaux, Luxembourg.
| |
Collapse
|
21
|
Haynes B, Murai J, Lee JM. Restored replication fork stabilization, a mechanism of PARP inhibitor resistance, can be overcome by cell cycle checkpoint inhibition. Cancer Treat Rev 2018; 71:1-7. [PMID: 30269007 DOI: 10.1016/j.ctrv.2018.09.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/06/2018] [Accepted: 09/09/2018] [Indexed: 02/06/2023]
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibition serves as a potent therapeutic option eliciting synthetic lethality in cancers harboring homologous recombination (HR) repair defects, such as BRCA mutations. However, the development of resistance to PARP inhibitors (PARPis) poses a clinical challenge. Restoration of HR competency is one of the many molecular factors contributing to PARPi resistance. Combination therapy with cell cycle checkpoint (ATR, CHK1, and WEE1) inhibitors is being investigated clinically in many cancers, particularly in ovarian cancer, to enhance the efficacy and circumvent resistance to PARPis. Ideally, inhibition of ATR, CHK1 and WEE1 proteins will abrogate G2 arrest and subsequent DNA repair via restored HR in PARPi-treated cells. Replication fork stabilization has recently been identified as a potential compensatory PARPi resistance mechanism, found in the absence of restored HR. ATR, CHK1, and WEE1 each possess different roles in replication fork stabilization, providing different mechanisms to consider when developing combination therapies to avoid continued development of drug resistance. This review examines the impact of ATR, CHK1, and WEE1 on replication fork stabilization. We also address the therapeutic potential for combining PARPis with cell cycle inhibitors and the possible consequence of combination therapies which do not adequately address both restored HR and replication fork stabilization as PARPi resistance mechanisms.
Collapse
Affiliation(s)
- Brittany Haynes
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - Junko Murai
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jung-Min Lee
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
22
|
You Y, Xu Z, Chen Y. Doxorubicin conjugated with a trastuzumab epitope and an MMP-2 sensitive peptide linker for the treatment of HER2-positive breast cancer. Drug Deliv 2018; 25:448-460. [PMID: 29405790 PMCID: PMC6058718 DOI: 10.1080/10717544.2018.1435746] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
HER2-positive breast cancer correlates with more aggressive tumor growth, a poorer prognosis and reduced overall survival. Currently, trastuzumab (Herceptin), which is an anti-HER2 antibody, is one of the key drugs. There is evidence indicating that conjugation of trastuzumab with chemotherapy drugs, such as doxorubicin (DOX), for multiple targets could be more effective. However, incomplete penetration into tumors has been noted for those conjugates. Compared to an antibody, peptides may represent an attractive alternative. For HER2, a similar potency has been observed for a 12-amino-acid anti-HER2 peptide mimetic YCDGFYACYMDV-NH2 (AHNP, disulfide-bridged) and full-length trastuzumab. Thus, a peptide, GPLGLAGDDYCDGFYACYMDV-NH2, which consists of AHNP and an MMP-2 cleavable linker GPLGLAGDD, was first designed, followed by conjugation with DOX via a glycine residue at the N-terminus to form a novel DOX-peptide conjugate MAHNP-DOX. Using HER2-positive human breast cancer cells BT474 and SKBR3 as in vitro model systems and nude mice with BT474 xenografts as an in vivo model, this conjugate was comprehensively characterized, and its efficacy was evaluated and compared with that of free DOX. As a result, MAHNP-DOX demonstrated a much lower in vitro IC50, and its in vivo extent of inhibition in mice was more evident. During this process, enzymatic cleavage of MAHNP-DOX is critical for its activation and cellular uptake. In addition, a synergistic response was observed after the combination of DOX and AHNP. This effect was probably due to the involvement of AHNP in the PI3K–AKT signaling pathway, which can be largely activated by DOX and leads to anti-apoptotic signals.
Collapse
Affiliation(s)
- Yiwen You
- a School of Pharmacy, Nanjing Medical University , Nanjing , China
| | - Zhiyuan Xu
- a School of Pharmacy, Nanjing Medical University , Nanjing , China
| | - Yun Chen
- a School of Pharmacy, Nanjing Medical University , Nanjing , China.,b State Key Laboratory of Reproductive Medicine , Nanjing , China
| |
Collapse
|
23
|
Minchom A, Aversa C, Lopez J. Dancing with the DNA damage response: next-generation anti-cancer therapeutic strategies. Ther Adv Med Oncol 2018; 10:1758835918786658. [PMID: 30023007 PMCID: PMC6047242 DOI: 10.1177/1758835918786658] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/08/2018] [Indexed: 01/01/2023] Open
Abstract
Maintenance of genomic stability is a critical determinant of cell survival and relies on the coordinated action of the DNA damage response (DDR), which orchestrates a network of cellular processes, including DNA replication, DNA repair and cell-cycle progression. In cancer, the critical balance between the loss of genomic stability in malignant cells and the DDR provides exciting therapeutic opportunities. Drugs targeting DDR pathways taking advantage of clinical synthetic lethality have already shown therapeutic benefit - for example, the PARP inhibitor olaparib has shown benefit in BRCA-mutant ovarian and breast cancer. Olaparib has also shown benefit in metastatic prostate cancer in DDR-defective patients, expanding the potential biomarker of response beyond BRCA. Other agents and combinations aiming to block the DDR while pushing damaged DNA through the cell cycle, including PARP, ATR, ATM, CHK and DNA-PK inhibitors, are in development. Emerging work is also uncovering how the DDR interacts intimately with the host immune response, including by activating the innate immune response, further suggesting that clinical applications together with immunotherapy may be beneficial. Here, we review recent considerations related to the DDR from a clinical standpoint, providing a framework to address future directions and clinical opportunities.
Collapse
Affiliation(s)
- Anna Minchom
- Drug Development Unit at Royal Marsden Hospital/ Institute of Cancer Research, Sutton, UK
| | - Caterina Aversa
- Drug Development Unit at Royal Marsden Hospital/ Institute of Cancer Research, Sutton, UK
| | - Juanita Lopez
- Drug Development Unit at Royal Marsden Hospital/Institute of Cancer Research, Downs Rd, Sutton, SM2 5PT, UK
| |
Collapse
|
24
|
Lindemann A, Takahashi H, Patel AA, Osman AA, Myers JN. Targeting the DNA Damage Response in OSCC with TP53 Mutations. J Dent Res 2018; 97:635-644. [PMID: 29489434 DOI: 10.1177/0022034518759068] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common type of oral cancer worldwide and in the United States. OSCC remains a major cause of morbidity and mortality in patients with head and neck cancers. Tobacco and alcohol consumption alone or with chewing betel nut are potential risk factors contributing to the high prevalence of OSCC. Multimodality therapies, including surgery, chemotherapy, biologic therapy, and radiotherapy, particularly intensity-modulated radiotherapy (IMRT), are the current treatments for OSCC patients. Despite recent advances in these treatment modalities, the overall survival remains poor over the past years. Recent data from whole-exome sequencing reveal that TP53 is commonly mutated in human papillomavirus-negative OSCC patients. Furthermore, these data stressed the importance of the TP53 gene in suppressing the development and progression of OSCC. Clinically, TP53 mutations are largely associated with poor survival and tumor resistance to radiotherapy and chemotherapy in OSCC patients, which makes the TP53 mutation status a potentially useful molecular marker prognostic and predictive of clinical response in these patients. Several forms of DNA damage have been shown to activate p53, including those generated by ionizing radiation and chemotherapy. The DNA damage stabilizes p53 in part via the DNA damage signaling pathway that involves sensor kinases, including ATM and ATR and effector kinases, such as Chk1/2 and Wee1, which leads to posttranscriptional regulation of a variety of genes involved in DNA repair, cell cycle control, apoptosis, and senescence. Here, we discuss the link of TP53 mutations with treatment outcome and survival in OSCC patients. We also provide evidence that small-molecule inhibitors of critical proteins that regulate DNA damage repair and replication stress during the cell cycle progression, as well as other molecules that restore wild-type p53 activity to mutant p53, can be exploited as novel therapeutic approaches for the treatment of OSCC patients bearing p53 mutant tumors.
Collapse
Affiliation(s)
- A Lindemann
- 1 Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - H Takahashi
- 1 Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A A Patel
- 1 Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A A Osman
- 1 Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J N Myers
- 1 Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
25
|
MK-8776, a novel chk1 kinase inhibitor, radiosensitizes p53-defective human tumor cells. Oncotarget 2018; 7:71660-71672. [PMID: 27690219 PMCID: PMC5342109 DOI: 10.18632/oncotarget.12311] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/20/2016] [Indexed: 12/20/2022] Open
Abstract
Radiotherapy is commonly used to treat a variety of solid tumors but improvements in the therapeutic ratio are sorely needed. The aim of this study was to assess the Chk1 kinase inhibitor, MK-8776, for its ability to radiosensitize human tumor cells. Cells derived from NSCLC and HNSCC cancers were tested for radiosensitization by MK-8776. The ability of MK-8776 to abrogate the radiation-induced G2 block was determined using flow cytometry. Effects on repair of radiation-induced DNA double strand breaks (DSBs) were determined on the basis of rad51, γ-H2AX and 53BP1 foci. Clonogenic survival analyses indicated that MK-8776 radiosensitized p53-defective tumor cells but not lines with wild-type p53. Abrogation of the G2 block was evident in both p53-defective cells and p53 wild-type lines indicating no correlation with radiosensitization. However, only p53-defective cells entered mitosis harboring unrepaired DSBs. MK-8776 appeared to inhibit repair of radiation-induced DSBs at early times after irradiation. A comparison of MK-8776 to the wee1 inhibitor, MK-1775, suggested both similarities and differences in their activities. In conclusion, MK-8776 radiosensitizes tumor cells by mechanisms that include abrogation of the G2 block and inhibition of DSB repair. Our findings support the clinical evaluation of MK-8776 in combination with radiation.
Collapse
|
26
|
BIGL: Biochemically Intuitive Generalized Loewe null model for prediction of the expected combined effect compatible with partial agonism and antagonism. Sci Rep 2017; 7:17935. [PMID: 29263342 PMCID: PMC5738392 DOI: 10.1038/s41598-017-18068-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/05/2017] [Indexed: 12/13/2022] Open
Abstract
Clinical efficacy regularly requires the combination of drugs. For an early estimation of the clinical value of (potentially many) combinations of pharmacologic compounds during discovery, the observed combination effect is typically compared to that expected under a null model. Mechanistic accuracy of that null model is not aspired to; to the contrary, combinations that deviate favorably from the model (and thereby disprove its accuracy) are prioritized. Arguably the most popular null model is the Loewe Additivity model, which conceptually maps any assay under study to a (virtual) single-step enzymatic reaction. It is easy-to-interpret and requires no other information than the concentration-response curves of the individual compounds. However, the original Loewe model cannot accommodate concentration-response curves with different maximal responses and, by consequence, combinations of an agonist with a partial or inverse agonist. We propose an extension, named Biochemically Intuitive Generalized Loewe (BIGL), that can address different maximal responses, while preserving the biochemical underpinning and interpretability of the original Loewe model. In addition, we formulate statistical tests for detecting synergy and antagonism, which allow for detecting statistically significant greater/lesser observed combined effects than expected from the null model. Finally, we demonstrate the novel method through application to several publicly available datasets.
Collapse
|
27
|
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: 210] [Impact Index Per Article: 30.0] [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
|
28
|
Carrassa L, Damia G. DNA damage response inhibitors: Mechanisms and potential applications in cancer therapy. Cancer Treat Rev 2017; 60:139-151. [PMID: 28961555 DOI: 10.1016/j.ctrv.2017.08.013] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/26/2017] [Accepted: 08/01/2017] [Indexed: 02/06/2023]
Abstract
Over the last decade the unravelling of the molecular mechanisms of the DNA damage response pathways and of the genomic landscape of human tumors have paved the road to new therapeutic approaches in oncology. It is now clear that tumors harbour defects in different DNA damage response steps, mainly signalling and repair, rendering them more dependent on the remaining pathways. We here focus on the proteins ATM, ATR, CHK1 and WEE1, reviewing their roles in the DNA damage response and as targets in cancer therapy. In the last decade specific inhibitors of these proteins have been designed, and their potential antineoplastic activity has been explored both in monotherapy strategies against tumors with specific defects (synthetic lethality approach) and in combination with radiotherapy or chemotherapeutic or molecular targeted agents. The preclinical and clinical evidence of antitumor activity of these inhibitors emanating from these research efforts will be critically reviewed. Lastly, the potential therapeutic feasibility of combining together such inhibitors with the aim to target particular subsets of tumors will be also discussed.
Collapse
Affiliation(s)
- Laura Carrassa
- Laboratory of Molecular Pharmacology, Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy.
| | - Giovanna Damia
- Laboratory of Molecular Pharmacology, Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy.
| |
Collapse
|
29
|
Narayan S, Jaiswal AS, Sharma R, Nawab A, Duckworth LV, Law BK, Zajac-Kaye M, George TJ, Sharma J, Sharma AK, Hromas RA. NSC30049 inhibits Chk1 pathway in 5-FU-resistant CRC bulk and stem cell populations. Oncotarget 2017; 8:57246-57264. [PMID: 28915668 PMCID: PMC5593639 DOI: 10.18632/oncotarget.19778] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/20/2017] [Indexed: 01/20/2023] Open
Abstract
The 5-fluorouracil (5-FU) treatment induces DNA damage and stalling of DNA replication forks. These stalled replication forks then collapse to form one sided double-strand breaks, leading to apoptosis. However, colorectal cancer (CRC) stem cells rapidly repair the stalled/collapsed replication forks and overcome treatment effects. Recent evidence suggests a critical role of checkpoint kinase 1 (Chk1) in preventing the replicative stress. Therefore, Chk1 kinase has been a target for developing mono or combination therapeutic agents. In the present study, we have identified a novel orphan molecule NSC30049 (NSC49L) that is effective alone, and in combination potentiates 5-FU-mediated growth inhibition of CRC heterogeneous bulk and FOLFOX-resistant cell lines in culture with minimal effect on normal colonic epithelial cells. It also inhibits the sphere forming activity of CRC stem cells, and decreases the expression levels of mRNAs of CRC stem cell marker genes. Results showed that NSC49L induces 5-FU-mediated S-phase cell cycle arrest due to increased load of DNA damage and increased γ-H2AX staining as a mechanism of cytotoxicity. The pharmacokinetic analysis showed a higher bioavailability of this compound, however, with a short plasma half-life. The drug is highly tolerated by animals with no pathological aberrations. Furthermore, NSC49L showed very potent activity in a HDTX model of CRC stem cell tumors either alone or in combination with 5-FU. Thus, NSC49L as a single agent or combined with 5-FU can be developed as a therapeutic agent by targeting the Chk1 pathway in 5-FU-resistant CRC heterogeneous bulk and CRC stem cell populations.
Collapse
Affiliation(s)
- Satya Narayan
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | - Aruna S. Jaiswal
- Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Ritika Sharma
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | - Akbar Nawab
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | - Lizette Vila Duckworth
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brian K. Law
- Department of Pharmacology and Experimental Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - Maria Zajac-Kaye
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | - Thomas J. George
- Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jay Sharma
- Celprogen, Inc., Torrance, CA 90503, USA
| | - Arun K. Sharma
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Robert A. Hromas
- Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
30
|
Na K, Li K, Sang T, Wu K, Wang Y, Wang X. Anticarcinogenic effects of water extract of sporoderm-broken spores of Ganoderma lucidum on colorectal cancer in vitro and in vivo. Int J Oncol 2017; 50:1541-1554. [PMID: 28358412 PMCID: PMC5403400 DOI: 10.3892/ijo.2017.3939] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 03/13/2017] [Indexed: 02/07/2023] Open
Abstract
Ganoderma lucidum (G. lucidum) polysaccharides (GLPs) have been used as traditional Chinese medicine for cancer prevention for many years. However, the mechanism by which GLP exerts its chemopreventive activities remains elusive. In addition, it is unclear whether sporoderm-broken spores of G. lucidum water extract (BSGLWE), which contains mainly GLPs, has anticancer effects on colorectal cancer. The present study investigated the anticancer effects and potential mechanisms of BSGLWE on colorectal cancer in vivo and in vitro. Our results showed that BSGLWE significantly inhibited colorectal cancer HCT116 cell viability in a time- and dose-dependent manner. Flow cytometry analysis indicated that BSGLWE disrupted cell cycle progression at G2/M phase via downregulation of cyclin B1 and cyclin A2, and upregulation of P21 at mRNA levels. Moreover, BSGLWE induced apoptosis by decreasing Bcl-2 and survivin at mRNA levels, and reduced Bcl-2, PARP, pro-caspase-3 and pro-caspase-9 at protein levels. Furthermore, BSGLWE suppressed tumor growth in vivo by regulating the expression of genes and proteins associated with cell cycle and apoptosis, which was further confirmed by a reduction of Ki67, PCNA, and Bcl-2 expression as determined by immunohistochemistry staining. NSAID activated gene-1 (NAG-1), a pro-apoptotic gene, was significantly upregulated in vivo and in vitro upon BSGLWE treatment at both mRNA and protein levels. In addition, the relative amounts of secreted NAG-1 in cell culture medium or serum of nude mice were all upregulated upon BSGLWE treatments, suggesting a role of NAG-1 in BSGLWE-induced anticolorectal cancer activity. This is the first study to show that BSGLWE inhibits colorectal cancer carcinogenesis through regulating genes responsible for cell proliferation, cell cycle and apoptosis cascades. These findings indicate that BSGLWE possesses chemopreventive potential in colorectal cancer which may serve as a promising anticancer agent for clinical applications.
Collapse
Affiliation(s)
- Kun Na
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Kang Li
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Tingting Sang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Kaikai Wu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Ying Wang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Xingya Wang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
31
|
Ghelli Luserna di Rora’ A, Iacobucci I, Martinelli G. The cell cycle checkpoint inhibitors in the treatment of leukemias. J Hematol Oncol 2017; 10:77. [PMID: 28356161 PMCID: PMC5371185 DOI: 10.1186/s13045-017-0443-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/15/2017] [Indexed: 01/25/2023] Open
Abstract
The inhibition of the DNA damage response (DDR) pathway in the treatment of cancers has recently reached an exciting stage with several cell cycle checkpoint inhibitors that are now being tested in several clinical trials in cancer patients. Although the great amount of pre-clinical and clinical data are from the solid tumor experience, only few studies have been done on leukemias using specific cell cycle checkpoint inhibitors. This review aims to summarize the most recent data found on the biological mechanisms of the response to DNA damages highlighting the role of the different elements of the DDR pathway in normal and cancer cells and focusing on the main genetic alteration or aberrant gene expression that has been found on acute and chronic leukemias. This review, for the first time, outlines the most important pre-clinical and clinical data available on the efficacy of cell cycle checkpoint inhibitors in single agent and in combination with different agents normally used for the treatment of acute and chronic leukemias.
Collapse
Affiliation(s)
| | - I. Iacobucci
- Department of Hematology and Medical Sciences “L. and A. Seràgnoli”, Bologna University, Bologna, Italy
- Present: Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - G. Martinelli
- Department of Hematology and Medical Sciences “L. and A. Seràgnoli”, Bologna University, Bologna, Italy
| |
Collapse
|
32
|
Lin AB, McNeely SC, Beckmann RP. Achieving Precision Death with Cell-Cycle Inhibitors that Target DNA Replication and Repair. Clin Cancer Res 2017; 23:3232-3240. [PMID: 28331049 DOI: 10.1158/1078-0432.ccr-16-0083] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/29/2016] [Accepted: 03/15/2017] [Indexed: 11/16/2022]
Abstract
All cancers are characterized by defects in the systems that ensure strict control of the cell cycle in normal tissues. The consequent excess tissue growth can be countered by drugs that halt cell division, and, indeed, the majority of chemotherapeutics developed during the last century work by disrupting processes essential for the cell cycle, particularly DNA synthesis, DNA replication, and chromatid segregation. In certain contexts, the efficacy of these classes of drugs can be impressive, but because they indiscriminately block the cell cycle of all actively dividing cells, their side effects severely constrain the dose and duration with which they can be administered, allowing both normal and malignant cells to escape complete growth arrest. Recent progress in understanding how cancers lose control of the cell cycle, coupled with comprehensive genomic profiling of human tumor biopsies, has shown that many cancers have mutations affecting various regulators and checkpoints that impinge on the core cell-cycle machinery. These defects introduce unique vulnerabilities that can be exploited by a next generation of drugs that promise improved therapeutic windows in patients whose tumors bear particular genomic aberrations, permitting increased dose intensity and efficacy. These developments, coupled with the success of new drugs targeting cell-cycle regulators, have led to a resurgence of interest in cell-cycle inhibitors. This review in particular focuses on the newer strategies that may facilitate better therapeutic targeting of drugs that inhibit the various components that safeguard the fidelity of the fundamental processes of DNA replication and repair. Clin Cancer Res; 23(13); 3232-40. ©2017 AACR.
Collapse
Affiliation(s)
- Aimee Bence Lin
- Early Phase Medical-Oncology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Samuel C McNeely
- Oncology Business Unit-Patient Tailoring, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Richard P Beckmann
- Oncology Translational Research, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana.
| |
Collapse
|
33
|
Leijen S, van Geel RM, Pavlick AC, Tibes R, Rosen L, Razak ARA, Lam R, Demuth T, Rose S, Lee MA, Freshwater T, Shumway S, Liang LW, Oza AM, Schellens JH, Shapiro GI. Phase I Study Evaluating WEE1 Inhibitor AZD1775 As Monotherapy and in Combination With Gemcitabine, Cisplatin, or Carboplatin in Patients With Advanced Solid Tumors. J Clin Oncol 2016; 34:4371-4380. [PMID: 27601554 PMCID: PMC7845944 DOI: 10.1200/jco.2016.67.5991] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Purpose AZD1775 is a WEE1 kinase inhibitor targeting G2 checkpoint control, preferentially sensitizing TP53-deficient tumor cells to DNA damage. This phase I study evaluated safety, tolerability, pharmacokinetics, and pharmacodynamics of oral AZD1775 as monotherapy or in combination with chemotherapy in patients with refractory solid tumors. Patients and Methods In part 1, patients received a single dose of AZD1775 followed by 14 days of observation. In part 2, patients received AZD1775 as a single dose (part 2A) or as five twice per day doses or two once per day doses (part 2B) in combination with one of the following chemotherapy agents: gemcitabine (1,000 mg/m2), cisplatin (75 mg/m2), or carboplatin (area under the curve, 5 mg/mL⋅min). Skin biopsies were collected for pharmacodynamic assessments. TP53 status was determined retrospectively in archival tumor tissue. Results Two hundred two patients were enrolled onto the study, including nine patients in part 1, 43 in part 2A (including eight rollover patients from part 1), and 158 in part 2B. AZD1775 monotherapy given as single dose was well tolerated, and the maximum-tolerated dose was not reached. In the combination regimens, the most common adverse events consisted of fatigue, nausea and vomiting, diarrhea, and hematologic toxicity. The maximum-tolerated doses and biologically effective doses were established for each combination. Target engagement, as a predefined 50% pCDK1 reduction in surrogate tissue, was observed in combination with cisplatin and carboplatin. Of 176 patients evaluable for efficacy, 94 (53%) had stable disease as best response, and 17 (10%) achieved a partial response. The response rate in TP53-mutated patients (n = 19) was 21% compared with 12% in TP53 wild-type patients (n = 33). Conclusion AZD1775 was safe and tolerable as a single agent and in combination with chemotherapy at doses associated with target engagement.
Collapse
Affiliation(s)
- Suzanne Leijen
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Robin M.J.M. van Geel
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Anna C. Pavlick
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Raoul Tibes
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Lee Rosen
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Albiruni R. Abdul Razak
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Raymond Lam
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Tim Demuth
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Shelonitda Rose
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Mark A. Lee
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Tomoko Freshwater
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Stuart Shumway
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Li Wen Liang
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Amit M. Oza
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Jan H.M. Schellens
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Geoffrey I. Shapiro
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| |
Collapse
|
34
|
Sakurikar N, Thompson R, Montano R, Eastman A. A subset of cancer cell lines is acutely sensitive to the Chk1 inhibitor MK-8776 as monotherapy due to CDK2 activation in S phase. Oncotarget 2016; 7:1380-94. [PMID: 26595527 PMCID: PMC4811467 DOI: 10.18632/oncotarget.6364] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/16/2015] [Indexed: 12/14/2022] Open
Abstract
DNA damage activates Checkpoint kinase 1 (Chk1) to halt cell cycle progression thereby preventing further DNA replication and mitosis until the damage has been repaired. Consequently, Chk1 inhibitors have emerged as promising anticancer therapeutics in combination with DNA damaging drugs, but their single agent activity also provides a novel approach that may be particularly effective in a subset of patients. From analysis of a large panel of cell lines, we demonstrate that 15% are very sensitive to the Chk1 inhibitor MK-8776. Upon inhibition of Chk1, sensitive cells rapidly accumulate DNA double-strand breaks in S phase in a CDK2- and cyclin A-dependent manner. In contrast, resistant cells can continue to grow for at least 7 days despite continued inhibition of Chk1. Resistance can be circumvented by inhibiting Wee1 kinase and thereby directly activating CDK2. Hence, sensitivity to Chk1 inhibition is regulated upstream of CDK2 and correlates with accumulation of CDC25A. We conclude that cells poorly tolerate CDK2 activity in S phase and that a major function of Chk1 is to ensure it remains inactive. Indeed, inhibitors of CDK1 and CDK2 arrest cells in G1 or G2, respectively, but do not prevent progression through S phase demonstrating that neither kinase is required for S phase progression. Inappropriate activation of CDK2 in S phase underlies the sensitivity of a subset of cell lines to Chk1 inhibitors, and this may provide a novel therapeutic opportunity for appropriately stratified patients.
Collapse
Affiliation(s)
- Nandini Sakurikar
- Department of Pharmacology and Toxicology, and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Ruth Thompson
- Department of Pharmacology and Toxicology, and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Ryan Montano
- Department of Pharmacology and Toxicology, and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Alan Eastman
- Department of Pharmacology and Toxicology, and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
35
|
Busch CJ, Kröger MS, Jensen J, Kriegs M, Gatzemeier F, Petersen C, Münscher A, Rothkamm K, Rieckmann T. G2-checkpoint targeting and radiosensitization of HPV/p16-positive HNSCC cells through the inhibition of Chk1 and Wee1. Radiother Oncol 2016; 122:260-266. [PMID: 27939202 DOI: 10.1016/j.radonc.2016.11.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/07/2016] [Accepted: 11/19/2016] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND PURPOSE HPV-positive HNSCC cells are characterized by radiosensitivity, inefficient DNA double-strand break repair and a profound and prolonged arrest in G2. Here we explored the effect of clinically relevant inhibitors of Chk1 and Wee1 to inhibit the radiation-induced G2-arrest in order to achieve further radiosensitization. MATERIAL AND METHODS Assessment of Chk1 activity by Western blot; assessment of cell cycle distribution by propidium iodide staining and flow cytometry; assessment of cell survival by colony formation assay. HPV+ HNSCC cell lines: UD-SCC-2, UM-SCC-47 and UPCI-SCC-154; Chk1 inhibitors: LY2603618, MK8776; Wee1 inhibitor: AZD1775. RESULTS Specific Chk1 inhibitors efficiently abrogated the radiation-induced G2-arrest and caused radiosensitization. Wee-inhibition by AZD1775 resulted in the activation of Chk1. This feedback mechanism is likely to counteract some of the effects of Wee1 inhibition but could be antagonized through the combined inhibition of both kinases. Combined inhibition was effective using profoundly reduced concentrations of both inhibitors and resulted in more efficient radiosensitization of the HPV-positive cell lines compared to p53 proficient normal human fibroblasts. CONCLUSIONS Specific Chk1 inhibitors as well as the combined inhibition of Chk1 and Wee1 radiosensitize HPV-positive HNSCC cells.
Collapse
Affiliation(s)
- Chia-Jung Busch
- Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Germany
| | - Marie Sophie Kröger
- Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Germany; Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Jana Jensen
- Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Germany; Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Malte Kriegs
- Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Fruzsina Gatzemeier
- Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Germany; Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Cordula Petersen
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Adrian Münscher
- Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Germany
| | - Kai Rothkamm
- Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Thorsten Rieckmann
- Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Germany; Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Germany.
| |
Collapse
|
36
|
Leijen S, van Geel RMJM, Sonke GS, de Jong D, Rosenberg EH, Marchetti S, Pluim D, van Werkhoven E, Rose S, Lee MA, Freshwater T, Beijnen JH, Schellens JHM. Phase II Study of WEE1 Inhibitor AZD1775 Plus Carboplatin in Patients With TP53-Mutated Ovarian Cancer Refractory or Resistant to First-Line Therapy Within 3 Months. J Clin Oncol 2016; 34:4354-4361. [PMID: 27998224 DOI: 10.1200/jco.2016.67.5942] [Citation(s) in RCA: 224] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose AZD1775 is a first-in-class, potent, and selective inhibitor of WEE1 with proof of chemopotentiation in p53-deficient tumors in preclinical models. In a phase I study, the maximum tolerated dose of AZD1775 in combination with carboplatin demonstrated target engagement. We conducted a proof-of-principle phase II study in patients with p53 tumor suppressor gene ( TP53)-mutated ovarian cancer refractory or resistant (< 3 months) to first-line platinum-based therapy to determine overall response rate, progression-free and overall survival, pharmacokinetics, and modulation of phosphorylated cyclin-dependent kinase (CDK1) in skin biopsies. Patients and Methods Patients were treated with carboplatin (area under the curve, 5 mg/mL⋅min) combined with AZD1775 225 mg orally twice daily over 2.5 days every 21-day cycle until disease progression. Results AZD1775 plus carboplatin demonstrated manageable toxicity; fatigue (87%), nausea (78%), thrombocytopenia (70%), diarrhea (70%), and vomiting (48%) were the most common adverse events. The most frequent grade 3 or 4 adverse events were thrombocytopenia (48%) and neutropenia (37%). Of 24 patients enrolled, 21 patients were evaluable for efficacy end points. The overall response rate was 43% (95% CI, 22% to 66%), including one patient (5%) with a prolonged complete response. Median progression-free and overall survival times were 5.3 months (95% CI, 2.3 to 9.0 months) and 12.6 months (95% CI, 4.9 to 19.7), respectively, with two patients with ongoing response for more than 31 and 42 months at data cutoff. Conclusion To our knowledge, this is the first report providing clinical proof that AZD1775 enhances carboplatin efficacy in TP53-mutated tumors. The encouraging antitumor activity observed in patients with TP53-mutated ovarian cancer who were refractory or resistant (< 3 months) to first-line therapy warrants further development.
Collapse
Affiliation(s)
- Suzanne Leijen
- Suzanne Leijen, Robin M.J.M. van Geel, Gabe S. Sonke, Daphne de Jong, Efraim H. Rosenberg, Serena Marchetti, Dick Pluim, Erik van Werkhoven, Jos H. Beijnen, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jos H. Beijnen and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; and Shelonitda Rose, Mark A. Lee, and Tomoko Freshwater, Merck, Kenilworth, NJ
| | - Robin M J M van Geel
- Suzanne Leijen, Robin M.J.M. van Geel, Gabe S. Sonke, Daphne de Jong, Efraim H. Rosenberg, Serena Marchetti, Dick Pluim, Erik van Werkhoven, Jos H. Beijnen, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jos H. Beijnen and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; and Shelonitda Rose, Mark A. Lee, and Tomoko Freshwater, Merck, Kenilworth, NJ
| | - Gabe S Sonke
- Suzanne Leijen, Robin M.J.M. van Geel, Gabe S. Sonke, Daphne de Jong, Efraim H. Rosenberg, Serena Marchetti, Dick Pluim, Erik van Werkhoven, Jos H. Beijnen, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jos H. Beijnen and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; and Shelonitda Rose, Mark A. Lee, and Tomoko Freshwater, Merck, Kenilworth, NJ
| | - Daphne de Jong
- Suzanne Leijen, Robin M.J.M. van Geel, Gabe S. Sonke, Daphne de Jong, Efraim H. Rosenberg, Serena Marchetti, Dick Pluim, Erik van Werkhoven, Jos H. Beijnen, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jos H. Beijnen and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; and Shelonitda Rose, Mark A. Lee, and Tomoko Freshwater, Merck, Kenilworth, NJ
| | - Efraim H Rosenberg
- Suzanne Leijen, Robin M.J.M. van Geel, Gabe S. Sonke, Daphne de Jong, Efraim H. Rosenberg, Serena Marchetti, Dick Pluim, Erik van Werkhoven, Jos H. Beijnen, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jos H. Beijnen and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; and Shelonitda Rose, Mark A. Lee, and Tomoko Freshwater, Merck, Kenilworth, NJ
| | - Serena Marchetti
- Suzanne Leijen, Robin M.J.M. van Geel, Gabe S. Sonke, Daphne de Jong, Efraim H. Rosenberg, Serena Marchetti, Dick Pluim, Erik van Werkhoven, Jos H. Beijnen, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jos H. Beijnen and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; and Shelonitda Rose, Mark A. Lee, and Tomoko Freshwater, Merck, Kenilworth, NJ
| | - Dick Pluim
- Suzanne Leijen, Robin M.J.M. van Geel, Gabe S. Sonke, Daphne de Jong, Efraim H. Rosenberg, Serena Marchetti, Dick Pluim, Erik van Werkhoven, Jos H. Beijnen, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jos H. Beijnen and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; and Shelonitda Rose, Mark A. Lee, and Tomoko Freshwater, Merck, Kenilworth, NJ
| | - Erik van Werkhoven
- Suzanne Leijen, Robin M.J.M. van Geel, Gabe S. Sonke, Daphne de Jong, Efraim H. Rosenberg, Serena Marchetti, Dick Pluim, Erik van Werkhoven, Jos H. Beijnen, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jos H. Beijnen and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; and Shelonitda Rose, Mark A. Lee, and Tomoko Freshwater, Merck, Kenilworth, NJ
| | - Shelonitda Rose
- Suzanne Leijen, Robin M.J.M. van Geel, Gabe S. Sonke, Daphne de Jong, Efraim H. Rosenberg, Serena Marchetti, Dick Pluim, Erik van Werkhoven, Jos H. Beijnen, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jos H. Beijnen and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; and Shelonitda Rose, Mark A. Lee, and Tomoko Freshwater, Merck, Kenilworth, NJ
| | - Mark A Lee
- Suzanne Leijen, Robin M.J.M. van Geel, Gabe S. Sonke, Daphne de Jong, Efraim H. Rosenberg, Serena Marchetti, Dick Pluim, Erik van Werkhoven, Jos H. Beijnen, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jos H. Beijnen and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; and Shelonitda Rose, Mark A. Lee, and Tomoko Freshwater, Merck, Kenilworth, NJ
| | - Tomoko Freshwater
- Suzanne Leijen, Robin M.J.M. van Geel, Gabe S. Sonke, Daphne de Jong, Efraim H. Rosenberg, Serena Marchetti, Dick Pluim, Erik van Werkhoven, Jos H. Beijnen, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jos H. Beijnen and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; and Shelonitda Rose, Mark A. Lee, and Tomoko Freshwater, Merck, Kenilworth, NJ
| | - Jos H Beijnen
- Suzanne Leijen, Robin M.J.M. van Geel, Gabe S. Sonke, Daphne de Jong, Efraim H. Rosenberg, Serena Marchetti, Dick Pluim, Erik van Werkhoven, Jos H. Beijnen, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jos H. Beijnen and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; and Shelonitda Rose, Mark A. Lee, and Tomoko Freshwater, Merck, Kenilworth, NJ
| | - Jan H M Schellens
- Suzanne Leijen, Robin M.J.M. van Geel, Gabe S. Sonke, Daphne de Jong, Efraim H. Rosenberg, Serena Marchetti, Dick Pluim, Erik van Werkhoven, Jos H. Beijnen, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jos H. Beijnen and Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; and Shelonitda Rose, Mark A. Lee, and Tomoko Freshwater, Merck, Kenilworth, NJ
| |
Collapse
|
37
|
Restelli V, Chilà R, Lupi M, Rinaldi A, Kwee I, Bertoni F, Damia G, Carrassa L. Characterization of a mantle cell lymphoma cell line resistant to the Chk1 inhibitor PF-00477736. Oncotarget 2016; 6:37229-40. [PMID: 26439697 PMCID: PMC4741926 DOI: 10.18632/oncotarget.5954] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/17/2015] [Indexed: 12/24/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell lymphoma characterized by the chromosomal translocation t(11;14) that leads to constitutive expression of cyclin D1, a master regulator of the G1-S phase. Chk1 inhibitors have been recently shown to be strongly effective as single agents in MCL. To investigate molecular mechanisms at the basis of Chk1 inhibitor activity, a MCL cell line resistant to the Chk1 inhibitor PF-00477736 (JEKO-1 R) was obtained and characterized. The JEKO-1 R cell line was cross resistant to another Chk1 inhibitor (AZD-7762) and to the Wee1 inhibitor MK-1775. It displayed a shorter doubling time than parental cell line, likely due to a faster S phase. Cyclin D1 expression levels were decreased in resistant cell line and its re-overexpression partially re-established PF-00477736 sensitivity. Gene expression profiling showed an enrichment in gene sets involved in pro-survival pathways in JEKO-1 R. Dasatinib treatment partly restored PF-00477736 sensitivity in resistant cells suggesting that the pharmacological interference of pro-survival pathways can overcome the resistance to Chk1 inhibitors. These data further corroborate the involvement of the t(11;14) in cellular sensitivity to Chk1 inhibitors, fostering the clinical testing of Chk1 inhibitors as single agents in MCL.
Collapse
Affiliation(s)
- Valentina Restelli
- Laboratory of Molecular Pharmacology and Laboratory of Cancer Pharmacology, Department of Oncology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Rosaria Chilà
- Laboratory of Molecular Pharmacology and Laboratory of Cancer Pharmacology, Department of Oncology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Monica Lupi
- Laboratory of Molecular Pharmacology and Laboratory of Cancer Pharmacology, Department of Oncology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Andrea Rinaldi
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland
| | - Ivo Kwee
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland.,Dalle Molle Institute for Artificial Intelligence (IDSIA), Manno, Switzerland.,Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Francesco Bertoni
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland.,Lymphoma Unit, IOSI Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Giovanna Damia
- Laboratory of Molecular Pharmacology and Laboratory of Cancer Pharmacology, Department of Oncology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Laura Carrassa
- Laboratory of Molecular Pharmacology and Laboratory of Cancer Pharmacology, Department of Oncology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| |
Collapse
|
38
|
Chemogenetic profiling identifies RAD17 as synthetically lethal with checkpoint kinase inhibition. Oncotarget 2016; 6:35755-69. [PMID: 26437225 PMCID: PMC4742139 DOI: 10.18632/oncotarget.5928] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 09/14/2015] [Indexed: 01/07/2023] Open
Abstract
Chemical inhibitors of the checkpoint kinases have shown promise in the treatment of cancer, yet their clinical utility may be limited by a lack of molecular biomarkers to identify specific patients most likely to respond to therapy. To this end, we screened 112 known tumor suppressor genes for synthetic lethal interactions with inhibitors of the CHEK1 and CHEK2 checkpoint kinases. We identified eight interactions, including the Replication Factor C (RFC)-related protein RAD17. Clonogenic assays in RAD17 knockdown cell lines identified a substantial shift in sensitivity to checkpoint kinase inhibition (3.5-fold) as compared to RAD17 wild-type. Additional evidence for this interaction was found in a large-scale functional shRNA screen of over 100 genotyped cancer cell lines, in which CHEK1/2 mutant cell lines were unexpectedly sensitive to RAD17 knockdown. This interaction was widely conserved, as we found that RAD17 interacts strongly with checkpoint kinases in the budding yeast Saccharomyces cerevisiae. In the setting of RAD17 knockdown, CHEK1/2 inhibition was found to be synergistic with inhibition of WEE1, another pharmacologically relevant checkpoint kinase. Accumulation of the DNA damage marker γH2AX following chemical inhibition or transient knockdown of CHEK1, CHEK2 or WEE1 was magnified by knockdown of RAD17. Taken together, our data suggest that CHEK1 or WEE1 inhibitors are likely to have greater clinical efficacy in tumors with RAD17 loss-of-function.
Collapse
|
39
|
Mak JPY, Man WY, Chow JPH, Ma HT, Poon RYC. Pharmacological inactivation of CHK1 and WEE1 induces mitotic catastrophe in nasopharyngeal carcinoma cells. Oncotarget 2016; 6:21074-84. [PMID: 26025928 PMCID: PMC4673251 DOI: 10.18632/oncotarget.4020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/09/2015] [Indexed: 12/23/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a rare but highly invasive cancer. As radiotherapy is the primary treatment for NPC, this offers a rationale to investigate if uncoupling the DNA damage responses can sensitize this cancer type. The G2 DNA damage checkpoint is controlled by a cascade of protein kinases: ATM/ATR, which phosphorylates CHK1/CHK2, which in turn phosphorylates WEE1. A number of small molecule inhibitors have been developed against these kinases as potential therapeutic agents. Here we demonstrated that compare to that in immortalized nasopharyngeal epithelial cells, ATR, CHK1, and WEE1 were overexpressed in NPC cell lines. Inhibitors of these kinases were unable to promote extensive mitotic catastrophe in ionizing radiation-treated NPC cells, indicating that they are not very effective radiosensitizer for this cancer. In the absence of prior irradiation, however, mitotic catastrophe could be induced with inhibitors against CHK1 (AZD7762) or WEE1 (MK-1775). NPC cells were more sensitive to WEE1 inactivation than nasopharyngeal epithelial cells. Targeting CHK1 and WEE1 together induced more extensive mitotic catastrophe than the individual components alone. Taken together, our results show that NPC cells depend on CHK1 and WEE1 activity for growth and that inhibitors of these kinases may serve as potential therapeutics for NPC.
Collapse
Affiliation(s)
- Joyce P Y Mak
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Wing Yu Man
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Jeremy P H Chow
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Hoi Tang Ma
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Randy Y C Poon
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| |
Collapse
|
40
|
Saini P, Li Y, Dobbelstein M. Wee1 is required to sustain ATR/Chk1 signaling upon replicative stress. Oncotarget 2016; 6:13072-87. [PMID: 25965828 PMCID: PMC4537000 DOI: 10.18632/oncotarget.3865] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/31/2015] [Indexed: 12/12/2022] Open
Abstract
The therapeutic efficacy of nucleoside analogues, e.g. gemcitabine, against cancer cells can be augmented by inhibitors of checkpoint kinases, including Wee1, ATR, and Chk1. We have compared the chemosensitizing effect of these inhibitors in cells derived from pancreatic cancer, a tumor entity where gemcitabine is part of the first-line therapeutic regimens, and in osteosarcoma-derived cells. As expected, all three inhibitors rendered cancer cells more sensitive to gemcitabine, but Wee1 inhibition proved to be particularly efficient in this context. Investigating the reasons for this potent sensitizing effect, we found that Wee1 inhibition or knockdown not only blocked Wee1 activity, but also reduced the activation of ATR/Chk1 in gemcitabine-treated cells. Combination of several inhibitors revealed that Wee1 inhibition requires Cyclin-dependent kinases 1 and 2 (Cdk1/2) and Polo-like kinase 1 (Plk1) to reduce ATR/Chk1 activity. Through activation of Cdks and Plk1, Wee1 inhibition reduces Claspin and CtIP levels, explaining the impairment in ATR/Chk1 activity. Taken together, these results confer a consistent signaling pathway reaching from Wee1 inhibition to impaired Chk1 activity, mechanistically dissecting how Wee1 inhibitors not only dysregulate cell cycle progression, but also enhance replicative stress and chemosensitivity towards nucleoside analogues.
Collapse
Affiliation(s)
- Priyanka Saini
- Institute of Molecular Oncology, Göttingen Centre of Molecular Biosciences (GZMB), Faculty of Medicine, University of Göttingen, Göttingen, Germany
| | - Yizhu Li
- Institute of Molecular Oncology, Göttingen Centre of Molecular Biosciences (GZMB), Faculty of Medicine, University of Göttingen, Göttingen, Germany
| | - Matthias Dobbelstein
- Institute of Molecular Oncology, Göttingen Centre of Molecular Biosciences (GZMB), Faculty of Medicine, University of Göttingen, Göttingen, Germany
| |
Collapse
|
41
|
Combined inhibition of Chk1 and Wee1 as a new therapeutic strategy for mantle cell lymphoma. Oncotarget 2016; 6:3394-408. [PMID: 25428911 PMCID: PMC4413661 DOI: 10.18632/oncotarget.2583] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 10/09/2014] [Indexed: 12/13/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive, incurable disease, characterized by a deregulated cell cycle. Chk1 and Wee1 are main regulators of cell cycle progression and recent data on solid tumors suggest that simultaneous inhibition of these proteins has a strong synergistic cytotoxic effect. The effects of a Chk1 inhibitor (PF-00477736) and a Wee1 inhibitor (MK-1775) have been herein investigated in a large panel of mature B-cell lymphoma cell lines. We found that MCL cells were the most sensitive to the Chk1 inhibitor PF-00477736 and Wee1 inhibitor MK-1775 as single agents. Possible involvement of the translocation t(11;14) in Chk1 inhibitor sensitivity was hypothesized. The combined inhibition of Chk1 and Wee1 was strongly synergistic in MCL cells, leading to deregulation of the cell cycle, with increased activity of CDK2 and CDK1, and activation of apoptosis. In vivo treatment with the drug combination of mice bearing JeKo-1 xenografts (MCL) had a marked antitumor effect with tumor regressions observed at non-toxic doses (best T/C%=0.54%). Gene expression profiling suggested effect on genes involved in apoptosis. The strong synergism observed by combining Chk1 and Wee1 inhibitors in preclinical models of MCL provides the rationale for testing this combination in the clinical setting.
Collapse
|
42
|
O'Neil J, Benita Y, Feldman I, Chenard M, Roberts B, Liu Y, Li J, Kral A, Lejnine S, Loboda A, Arthur W, Cristescu R, Haines BB, Winter C, Zhang T, Bloecher A, Shumway SD. An Unbiased Oncology Compound Screen to Identify Novel Combination Strategies. Mol Cancer Ther 2016; 15:1155-62. [PMID: 26983881 DOI: 10.1158/1535-7163.mct-15-0843] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 03/07/2016] [Indexed: 12/15/2022]
Abstract
Combination drug therapy is a widely used paradigm for managing numerous human malignancies. In cancer treatment, additive and/or synergistic drug combinations can convert weakly efficacious monotherapies into regimens that produce robust antitumor activity. This can be explained in part through pathway interdependencies that are critical for cancer cell proliferation and survival. However, identification of the various interdependencies is difficult due to the complex molecular circuitry that underlies tumor development and progression. Here, we present a high-throughput platform that allows for an unbiased identification of synergistic and efficacious drug combinations. In a screen of 22,737 experiments of 583 doublet combinations in 39 diverse cancer cell lines using a 4 by 4 dosing regimen, both well-known and novel synergistic and efficacious combinations were identified. Here, we present an example of one such novel combination, a Wee1 inhibitor (AZD1775) and an mTOR inhibitor (ridaforolimus), and demonstrate that the combination potently and synergistically inhibits cancer cell growth in vitro and in vivo This approach has identified novel combinations that would be difficult to reliably predict based purely on our current understanding of cancer cell biology. Mol Cancer Ther; 15(6); 1155-62. ©2016 AACR.
Collapse
Affiliation(s)
| | - Yair Benita
- Merck Research Laboratories, Boston, Massachusetts
| | - Igor Feldman
- Merck Research Laboratories, Boston, Massachusetts
| | | | | | - Yaping Liu
- Merck Research Laboratories, North Wales, Pennsylvania
| | - Jing Li
- Merck Research Laboratories, North Wales, Pennsylvania
| | - Astrid Kral
- Merck Research Laboratories, Boston, Massachusetts
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Friedman AA, Amzallag A, Pruteanu-Malinici I, Baniya S, Cooper ZA, Piris A, Hargreaves L, Igras V, Frederick DT, Lawrence DP, Haber DA, Flaherty KT, Wargo JA, Ramaswamy S, Benes CH, Fisher DE. Landscape of Targeted Anti-Cancer Drug Synergies in Melanoma Identifies a Novel BRAF-VEGFR/PDGFR Combination Treatment. PLoS One 2015; 10:e0140310. [PMID: 26461489 PMCID: PMC4604168 DOI: 10.1371/journal.pone.0140310] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 09/24/2015] [Indexed: 12/12/2022] Open
Abstract
A newer generation of anti-cancer drugs targeting underlying somatic genetic driver events have resulted in high single-agent or single-pathway response rates in selected patients, but few patients achieve complete responses and a sizeable fraction of patients relapse within a year. Thus, there is a pressing need for identification of combinations of targeted agents which induce more complete responses and prevent disease progression. We describe the results of a combination screen of an unprecedented scale in mammalian cells performed using a collection of targeted, clinically tractable agents across a large panel of melanoma cell lines. We find that even the most synergistic drug pairs are effective only in a discrete number of cell lines, underlying a strong context dependency for synergy, with strong, widespread synergies often corresponding to non-specific or off-target drug effects such as multidrug resistance protein 1 (MDR1) transporter inhibition. We identified drugs sensitizing cell lines that are BRAFV600E mutant but intrinsically resistant to BRAF inhibitor PLX4720, including the vascular endothelial growth factor receptor/kinase insert domain receptor (VEGFR/KDR) and platelet derived growth factor receptor (PDGFR) family inhibitor cediranib. The combination of cediranib and PLX4720 induced apoptosis in vitro and tumor regression in animal models. This synergistic interaction is likely due to engagement of multiple receptor tyrosine kinases (RTKs), demonstrating the potential of drug- rather than gene-specific combination discovery approaches. Patients with elevated biopsy KDR expression showed decreased progression free survival in trials of mitogen-activated protein kinase (MAPK) kinase pathway inhibitors. Thus, high-throughput unbiased screening of targeted drug combinations, with appropriate library selection and mechanistic follow-up, can yield clinically-actionable drug combinations.
Collapse
Affiliation(s)
- Adam A. Friedman
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Arnaud Amzallag
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Iulian Pruteanu-Malinici
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Subash Baniya
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Zachary A. Cooper
- Department of Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
- Department of Genomic Medicine, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Adriano Piris
- Division of Dermatopathology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Leeza Hargreaves
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Vivien Igras
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Dennie T. Frederick
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Donald P. Lawrence
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Daniel A. Haber
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Keith T. Flaherty
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jennifer A. Wargo
- Department of Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
- Department of Genomic Medicine, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Sridhar Ramaswamy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Cyril H. Benes
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - David E. Fisher
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| |
Collapse
|
44
|
Manic G, Obrist F, Sistigu A, Vitale I. Trial Watch: Targeting ATM-CHK2 and ATR-CHK1 pathways for anticancer therapy. Mol Cell Oncol 2015; 2:e1012976. [PMID: 27308506 PMCID: PMC4905354 DOI: 10.1080/23723556.2015.1012976] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/25/2015] [Accepted: 01/26/2015] [Indexed: 02/08/2023]
Abstract
The ataxia telangiectasia mutated serine/threonine kinase (ATM)/checkpoint kinase 2 (CHEK2, best known as CHK2) and the ATM and Rad3-related serine/threonine kinase (ATR)/CHEK1 (best known as CHK1) cascades are the 2 major signaling pathways driving the DNA damage response (DDR), a network of processes crucial for the preservation of genomic stability that act as a barrier against tumorigenesis and tumor progression. Mutations and/or deletions of ATM and/or CHK2 are frequently found in tumors and predispose to cancer development. In contrast, the ATR-CHK1 pathway is often upregulated in neoplasms and is believed to promote tumor growth, although some evidence indicates that ATR and CHK1 may also behave as haploinsufficient oncosuppressors, at least in a specific genetic background. Inactivation of the ATM-CHK2 and ATR-CHK1 pathways efficiently sensitizes malignant cells to radiotherapy and chemotherapy. Moreover, ATR and CHK1 inhibitors selectively kill tumor cells that present high levels of replication stress, have a deficiency in p53 (or other DDR players), or upregulate the ATR-CHK1 module. Despite promising preclinical results, the clinical activity of ATM, ATR, CHK1, and CHK2 inhibitors, alone or in combination with other therapeutics, has not yet been fully demonstrated. In this Trial Watch, we give an overview of the roles of the ATM-CHK2 and ATR-CHK1 pathways in cancer initiation and progression, and summarize the results of clinical studies aimed at assessing the safety and therapeutic profile of regimens based on inhibitors of ATR and CHK1, the only 2 classes of compounds that have so far entered clinics.
Collapse
Affiliation(s)
| | - Florine Obrist
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
- INSERM, UMRS1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | | | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
- Department of Biology, University of Rome “TorVergata”; Rome, Italy
| |
Collapse
|
45
|
Mak JPY, Man WY, Ma HT, Poon RYC. Pharmacological targeting the ATR-CHK1-WEE1 axis involves balancing cell growth stimulation and apoptosis. Oncotarget 2015; 5:10546-57. [PMID: 25301733 PMCID: PMC4279392 DOI: 10.18632/oncotarget.2508] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 09/24/2014] [Indexed: 02/04/2023] Open
Abstract
The ATR–CHK1–WEE1 kinase cascade's functions in the DNA damage checkpoints are well established. Moreover, its roles in the unperturbed cell cycle are also increasingly being recognized. In this connection, a number of small-molecule inhibitors of ATR, CHK1, and WEE1 are being evaluated in clinical trials. Understanding precisely how cells respond to different concentrations of inhibitors is therefore of paramount importance and has broad clinical implications. Here we present evidence that in the absence of DNA damage, pharmacological inactivation of ATR was less effective in inducing mitotic catastrophe than inhibition of WEE1 and CHK1. Small-molecule inhibitors of CHK1 (AZD7762) or WEE1 (MK-1775) induced mitotic catastrophe, as characterized by dephosphorylation of CDK1Tyr15, phosphorylation of histone H3Ser10, and apoptosis. Unexpectedly, partial inhibition of WEE1 and CHK1 had the opposite effect of accelerating the cell cycle without inducing apoptosis, thereby increasing the overall cell proliferation. This was also corroborated by the finding that cell proliferation was enhanced by kinase-inactive versions of WEE1. We demonstrated that these potential limitations of the inhibitors could be overcome by targeting more than one components of the ATR–CHK1–WEE1 simultaneously. These observations reveal insights into the complex responses to pharmacological inactivation of the ATR–CHK1–WEE1 axis.
Collapse
Affiliation(s)
- Joyce P Y Mak
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Wing Yu Man
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Hoi Tang Ma
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Randy Y C Poon
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| |
Collapse
|
46
|
Magnussen GI, Emilsen E, Giller Fleten K, Engesæter B, Nähse-Kumpf V, Fjær R, Slipicevic A, Flørenes VA. Combined inhibition of the cell cycle related proteins Wee1 and Chk1/2 induces synergistic anti-cancer effect in melanoma. BMC Cancer 2015; 15:462. [PMID: 26054341 PMCID: PMC4460948 DOI: 10.1186/s12885-015-1474-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 05/26/2015] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Malignant melanoma has an increasing incidence rate and the metastatic disease is notoriously resistant to standard chemotherapy. Loss of cell cycle checkpoints is frequently found in many cancer types and makes the cells reliant on compensatory mechanisms to control progression. This feature may be exploited in therapy, and kinases involved in checkpoint regulation, such as Wee1 and Chk1/2, have thus become attractive therapeutic targets. METHODS In the present study we combined a Wee1 inhibitor (MK1775) with Chk1/2 inhibitor (AZD7762) in malignant melanoma cell lines grown in vitro (2D and 3D cultures) and in xenografts models. RESULTS Our in vitro studies showed that combined inhibition of Wee1 and Chk1/2 synergistically decreased viability and increased apoptosis (cleavage of caspase 3 and PARP), which may be explained by accumulation of DNA-damage (increased expression of γ-H2A.X)--and premature mitosis of S-phase cells. Compared to either inhibitor used as single agents, combined treatment reduced spheroid growth and led to greater tumour growth inhibition in melanoma xenografts. CONCLUSIONS These data provide a rationale for further evaluation of the combination of Wee1 and Chk1/2 inhibitors in malignant melanoma.
Collapse
Affiliation(s)
- Gry Irene Magnussen
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, N-0310, Oslo, Norway.
| | - Elisabeth Emilsen
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, N-0310, Oslo, Norway.
| | - Karianne Giller Fleten
- Department of Tumour Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo, Norway.
| | - Birgit Engesæter
- Department of Tumour Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo, Norway.
| | - Viola Nähse-Kumpf
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo, Norway.
| | - Roar Fjær
- Department of Medical Genetics, Ullevål University Hospital, Oslo, Norway.
| | - Ana Slipicevic
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, N-0310, Oslo, Norway.
| | - Vivi Ann Flørenes
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, N-0310, Oslo, Norway.
| |
Collapse
|
47
|
Liu D, Wu C, Jiao Y, Hou L, Lu D, Zheng H, Chen C, Qian J, Fei K, Su B. WEE1 kinase polymorphism as a predictive biomarker for efficacy of platinum-gemcitabine doublet chemotherapy in advanced non-small cell lung cancer patients. Sci Rep 2015; 5:11114. [PMID: 26057002 PMCID: PMC4460872 DOI: 10.1038/srep11114] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 05/11/2015] [Indexed: 12/31/2022] Open
Abstract
DNA-damaging agents are commonly used for first-line chemotherapy of advanced non-small cell lung cancer (NSCLC). As a G2/M checkpoint kinase, Wee1 can phosphorylate CDC2-tyr15 and induce G2/M cell cycle arrest in response to DNA damage. The correlation of WEE1 polymorphisms to the efficacy of chemotherapy was tested in 663 advanced NSCLC patients. WEE1 rs3910384 genotype correlated to overall survival (OS) and progress-free survival (PFS) of NSCLC patients treated with platinum-based chemotherapy. Sub-group analysis revealed that rs3910384 was particularly associated with the efficacy of doublet chemotherapy combining two DNA-damaging agents, i.e. platinum and gemcitabine. NSCLC patients with the WEE1 rs3910384 G/G homozygote genotype showed 13.5 months extended OS, 3.2 months extended PFS, and a 274% relative increase in their 3-year survival rate (from 7.4% to 27.7%) compared to the A/A+A/G genotype after treatment with platinum-gemcitabine regimen. This finding was reproduced in the validation cohort. We utilized a luciferase reporter assay and Electrophoretic Mobility Shift Assay (EMSA) to demonstrate that rs3910384-linked WEE1 promoter haplotype can mediate allele-specific transcriptional binding and WEE1 expression in DNA damage response. In conclusion, the WEE1 rs3910384 G/G homozygote genotype can be used as a selective biomarker for NSCLC patients to indicate treatment with platinum and gemcitabine regimen.
Collapse
Affiliation(s)
| | | | | | | | - Daru Lu
- Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Hui Zheng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine
| | - Ji Qian
- Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Ke Fei
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine
| | | |
Collapse
|
48
|
Dobbelstein M, Sørensen CS. Exploiting replicative stress to treat cancer. Nat Rev Drug Discov 2015; 14:405-23. [PMID: 25953507 DOI: 10.1038/nrd4553] [Citation(s) in RCA: 218] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
DNA replication in cancer cells is accompanied by stalling and collapse of the replication fork and signalling in response to DNA damage and/or premature mitosis; these processes are collectively known as 'replicative stress'. Progress is being made to increase our understanding of the mechanisms that govern replicative stress, thus providing ample opportunities to enhance replicative stress for therapeutic purposes. Rather than trying to halt cell cycle progression, cancer therapeutics could aim to increase replicative stress by further loosening the checkpoints that remain available to cancer cells and ultimately inducing the catastrophic failure of proliferative machineries. In this Review, we outline current and future approaches to achieve this, emphasizing the combination of conventional chemotherapy with targeted approaches.
Collapse
Affiliation(s)
- Matthias Dobbelstein
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences, Ernst Caspari Haus, University of Göttingen, 37077 Göttingen, Germany
| | | |
Collapse
|
49
|
Bose P, Grant S. Rational Combinations of Targeted Agents in AML. J Clin Med 2015; 4:634-664. [PMID: 26113989 PMCID: PMC4470160 DOI: 10.3390/jcm4040634] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 01/06/2015] [Indexed: 12/20/2022] Open
Abstract
Despite modest improvements in survival over the last several decades, the treatment of AML continues to present a formidable challenge. Most patients are elderly, and these individuals, as well as those with secondary, therapy-related, or relapsed/refractory AML, are particularly difficult to treat, owing to both aggressive disease biology and the high toxicity of current chemotherapeutic regimens. It has become increasingly apparent in recent years that coordinated interruption of cooperative survival signaling pathways in malignant cells is necessary for optimal therapeutic results. The modest efficacy of monotherapy with both cytotoxic and targeted agents in AML testifies to this. As the complex biology of AML continues to be elucidated, many “synthetic lethal” strategies involving rational combinations of targeted agents have been developed. Unfortunately, relatively few of these have been tested clinically, although there is growing interest in this area. In this article, the preclinical and, where available, clinical data on some of the most promising rational combinations of targeted agents in AML are summarized. While new molecules should continue to be combined with conventional genotoxic drugs of proven efficacy, there is perhaps a need to rethink traditional philosophies of clinical trial development and regulatory approval with a focus on mechanism-based, synergistic strategies.
Collapse
Affiliation(s)
- Prithviraj Bose
- Department of Internal Medicine, Virginia Commonwealth University and VCU Massey Cancer Center Center, 1201 E Marshall St, MMEC 11-213, P.O. Box 980070, Richmond, VA 23298, USA; E-Mail:
| | - Steven Grant
- Departments of Internal Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, Human and Molecular Genetics and the Institute for Molecular Medicine, Virginia Commonwealth University and VCU Massey Cancer Center, 401 College St, P.O. Box 980035, Richmond, VA 23298, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-804-828-5211; Fax: +1-804-628-5920
| |
Collapse
|
50
|
Aarts M, Bajrami I, Herrera-Abreu MT, Elliott R, Brough R, Ashworth A, Lord CJ, Turner NC. Functional Genetic Screen Identifies Increased Sensitivity to WEE1 Inhibition in Cells with Defects in Fanconi Anemia and HR Pathways. Mol Cancer Ther 2015; 14:865-76. [PMID: 25673822 PMCID: PMC6485454 DOI: 10.1158/1535-7163.mct-14-0845] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/04/2015] [Indexed: 01/11/2023]
Abstract
WEE1 kinase regulates CDK1 and CDK2 activity to facilitate DNA replication during S-phase and to prevent unscheduled entry into mitosis. WEE1 inhibitors synergize with DNA-damaging agents that arrest cells in S-phase by triggering direct mitotic entry without completing DNA synthesis, resulting in catastrophic chromosome fragmentation and apoptosis. Here, we investigated how WEE1 inhibition could be best exploited for cancer therapy by performing a functional genetic screen to identify novel determinants of sensitivity to WEE1 inhibition. Inhibition of kinases that regulate CDK activity, CHK1 and MYT1, synergized with WEE1 inhibition through both increased replication stress and forced mitotic entry of S-phase cells. Loss of multiple components of the Fanconi anemia (FA) and homologous recombination (HR) pathways, in particular DNA helicases, sensitized to WEE1 inhibition. Silencing of FA/HR genes resulted in excessive replication stress and nucleotide depletion following WEE1 inhibition, which ultimately led to increased unscheduled mitotic entry. Our results suggest that cancers with defects in FA and HR pathways may be targeted by WEE1 inhibition, providing a basis for a novel synthetic lethal strategy for cancers harboring FA/HR defects.
Collapse
Affiliation(s)
- Marieke Aarts
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Ilirjana Bajrami
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Maria T Herrera-Abreu
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Richard Elliott
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Rachel Brough
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Alan Ashworth
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Christopher J Lord
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Nicholas C Turner
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom. Breast Unit, Royal Marsden Hospital, London, United Kingdom.
| |
Collapse
|