1
|
Asfa SS, Arshinchi Bonab R, Önder O, Uça Apaydın M, Döşeme H, Küçük C, Georgakilas AG, Stadler BM, Logotheti S, Kale S, Pavlopoulou A. Computer-Aided Identification and Design of Ligands for Multi-Targeting Inhibition of a Molecular Acute Myeloid Leukemia Network. Cancers (Basel) 2024; 16:3607. [PMID: 39518047 PMCID: PMC11544916 DOI: 10.3390/cancers16213607] [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: 08/29/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Acute myeloid leukemia (AML) is characterized by therapeutic failure and long-term risk for disease relapses. As several therapeutic targets participate in networks, they can rewire to eventually evade single-target drugs. Hence, multi-targeting approaches are considered on the expectation that interference with many different components could synergistically hinder activation of alternative pathways and demolish the network one-off, leading to complete disease remission. METHODS Herein, we established a network-based, computer-aided approach for the rational design of drug combinations and de novo agents that interact with many AML network components simultaneously. RESULTS A reconstructed AML network guided the selection of suitable protein hubs and corresponding multi-targeting strategies. For proteins responsive to existing drugs, a greedy algorithm identified the minimum amount of compounds targeting the maximum number of hubs. We predicted permissible combinations of amiodarone, artenimol, fostamatinib, ponatinib, procaine, and vismodegib that interfere with 3-8 hubs, and we elucidated the pharmacological mode of action of procaine on DNMT3A. For proteins that do not respond to any approved drugs, namely cyclins A1, D2, and E1, we used structure-based de novo drug design to generate a novel triple-targeting compound of the chemical formula C15H15NO5, with favorable pharmacological and drug-like properties. CONCLUSIONS Overall, by integrating network and structural pharmacology with molecular modeling, we determined two complementary strategies with the potential to annihilate the AML network, one in the form of repurposable drug combinations and the other as a de novo synthesized triple-targeting agent. These target-drug interactions could be prioritized for preclinical and clinical testing toward precision medicine for AML.
Collapse
Affiliation(s)
- Seyedeh Sadaf Asfa
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
- Department of Pharmacology and Therapeutics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R3E 0W2, Canada
| | - Reza Arshinchi Bonab
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
- Department of Pharmacology and Therapeutics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R3E 0W2, Canada
| | - Onur Önder
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
| | - Merve Uça Apaydın
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
| | - Hatice Döşeme
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
| | - Can Küçük
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylül University, 35330 Balçova, İzmir, Türkiye;
| | - Alexandros G. Georgakilas
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou Campous, 15780 Athens, Greece;
| | - Bernhard M. Stadler
- Technische Hochschule Nürnberg, Faculty of Applied Chemistry, 90489 Nuremberg, Germany;
| | - Stella Logotheti
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Seyit Kale
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Department of Biophysics, Faculty of Medicine, Izmir Katip Çelebi University, 35330 Çiğli, İzmir, Türkiye
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
| |
Collapse
|
2
|
Stewart A, Song J, Pickard L, Muggiolu G, Sauvaigo S, Brandon ADH, Raynaud F, Banerji U. Characterizing functional DNA damage and response caused by the combination of CHK1 and WEE1 inhibitors in ovarian and breast cancer models. BJC REPORTS 2024; 2:27. [PMID: 39516567 PMCID: PMC11523970 DOI: 10.1038/s44276-024-00048-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND We proposed to quantify reduction of functional DNA damage response (DDR) mechanisms caused by the combination of CHK1 and WEE1 inhibitors. METHODS Survival of cells and tumor growth in-vitro and in-vivo caused by the combination of the CHK1 inhibitor SRA737 and the WEE1 inhibitor adavosertib was studied in OVCAR3 and MDA-MB 436 cells. Functional DNA damage was quantified using in vitro cell free DNA assays. RESULTS The combination of SRA737 and adavosertib caused significant reduction of survival of cells and DNA damage in-vitro and growth inhibition in-vivo. Studies using functional DDR assays found significant changes in the functional capacity of OVCAR3 but not MDA-MB 436 cells to repair DNA damage using multiple mechanisms including intra strand cross link repair, nucleotide excision repair, homologous recombination and non-homologous end joining. This study, for the first time provides a mechanistic insight into differences in the reduction in functional capacity of cells to repair DNA when exposed to CHK1 and WEE1 inhibitors. CONCLUSION The combination of the CHK1 inhibitor SRA737 and WEE1 inhibitor adavosertib causes growth inhibition in-vitro and in-vivo, but differential functional inhibition of DDR in the models studied.
Collapse
Affiliation(s)
| | - Jiin Song
- The Institute of Cancer Research, London, UK
| | | | | | | | | | | | | |
Collapse
|
3
|
De Mel S, Lee AR, Tan JHI, Tan RZY, Poon LM, Chan E, Lee J, Chee YL, Lakshminarasappa SR, Jaynes PW, Jeyasekharan AD. Targeting the DNA damage response in hematological malignancies. Front Oncol 2024; 14:1307839. [PMID: 38347838 PMCID: PMC10859481 DOI: 10.3389/fonc.2024.1307839] [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: 10/05/2023] [Accepted: 01/03/2024] [Indexed: 02/15/2024] Open
Abstract
Deregulation of the DNA damage response (DDR) plays a critical role in the pathogenesis and progression of many cancers. The dependency of certain cancers on DDR pathways has enabled exploitation of such through synthetically lethal relationships e.g., Poly ADP-Ribose Polymerase (PARP) inhibitors for BRCA deficient ovarian cancers. Though lagging behind that of solid cancers, DDR inhibitors (DDRi) are being clinically developed for haematological cancers. Furthermore, a high proliferative index characterize many such cancers, suggesting a rationale for combinatorial strategies targeting DDR and replicative stress. In this review, we summarize pre-clinical and clinical data on DDR inhibition in haematological malignancies and highlight distinct haematological cancer subtypes with activity of DDR agents as single agents or in combination with chemotherapeutics and targeted agents. We aim to provide a framework to guide the design of future clinical trials involving haematological cancers for this important class of drugs.
Collapse
Affiliation(s)
- Sanjay De Mel
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Ainsley Ryan Lee
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Joelle Hwee Inn Tan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Rachel Zi Yi Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Li Mei Poon
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, National University Health System, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Esther Chan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, National University Health System, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Joanne Lee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, National University Health System, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Yen Lin Chee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, National University Health System, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Satish R. Lakshminarasappa
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Patrick William Jaynes
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Anand D. Jeyasekharan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| |
Collapse
|
4
|
Tan J, Sun X, Zhao H, Guan H, Gao S, Zhou P. Double-strand DNA break repair: molecular mechanisms and therapeutic targets. MedComm (Beijing) 2023; 4:e388. [PMID: 37808268 PMCID: PMC10556206 DOI: 10.1002/mco2.388] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/29/2023] [Accepted: 09/08/2023] [Indexed: 10/10/2023] Open
Abstract
Double-strand break (DSB), a significant DNA damage brought on by ionizing radiation, acts as an initiating signal in tumor radiotherapy, causing cancer cells death. The two primary pathways for DNA DSB repair in mammalian cells are nonhomologous end joining (NHEJ) and homologous recombination (HR), which cooperate and compete with one another to achieve effective repair. The DSB repair mechanism depends on numerous regulatory variables. DSB recognition and the recruitment of DNA repair components, for instance, depend on the MRE11-RAD50-NBS1 (MRN) complex and the Ku70/80 heterodimer/DNA-PKcs (DNA-PK) complex, whose control is crucial in determining the DSB repair pathway choice and efficiency of HR and NHEJ. In-depth elucidation on the DSB repair pathway's molecular mechanisms has greatly facilitated for creation of repair proteins or pathways-specific inhibitors to advance precise cancer therapy and boost the effectiveness of cancer radiotherapy. The architectures, roles, molecular processes, and inhibitors of significant target proteins in the DSB repair pathways are reviewed in this article. The strategy and application in cancer therapy are also discussed based on the advancement of inhibitors targeted DSB damage response and repair proteins.
Collapse
Affiliation(s)
- Jinpeng Tan
- Hengyang Medical CollegeUniversity of South ChinaHengyangHunan ProvinceChina
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Xingyao Sun
- Hengyang Medical CollegeUniversity of South ChinaHengyangHunan ProvinceChina
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Hongling Zhao
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Hua Guan
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Shanshan Gao
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Ping‐Kun Zhou
- Hengyang Medical CollegeUniversity of South ChinaHengyangHunan ProvinceChina
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| |
Collapse
|
5
|
Zhang F, Chen L. Molecular Threat of Splicing Factor Mutations to Myeloid Malignancies and Potential Therapeutic Modulations. Biomedicines 2022; 10:biomedicines10081972. [PMID: 36009519 PMCID: PMC9405558 DOI: 10.3390/biomedicines10081972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/21/2022] Open
Abstract
Splicing factors are frequently mutated in myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). These mutations are presumed to contribute to oncogenic transformation, but the underlying mechanisms remain incompletely understood. While no specific treatment option is available for MDS/AML patients with spliceosome mutations, novel targeting strategies are actively explored, leading to clinical trials of small molecule inhibitors that target the spliceosome, DNA damage response pathway, and immune response pathway. Here, we review recent progress in mechanistic understanding of splicing factor mutations promoting disease progression and summarize potential therapeutic strategies, which, if successful, would provide clinical benefit to patients carrying splicing factor mutations.
Collapse
|
6
|
Wang LW, Jiang S, Yuan YH, Duan J, Mao ND, Hui Z, Bai R, Xie T, Ye XY. Recent Advances in Synergistic Antitumor Effects Exploited from the Inhibition of Ataxia Telangiectasia and RAD3-Related Protein Kinase (ATR). Molecules 2022; 27:molecules27082491. [PMID: 35458687 PMCID: PMC9029554 DOI: 10.3390/molecules27082491] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/27/2022] [Accepted: 04/05/2022] [Indexed: 02/04/2023] Open
Abstract
As one of the key phosphatidylinositol 3-kinase-related kinases (PIKKs) family members, ataxia telangiectasia and RAD3-related protein kinase (ATR) is crucial in maintaining mammalian cell genomic integrity in DNA damage response (DDR) and repair pathways. Dysregulation of ATR has been found across different cancer types. In recent years, the inhibition of ATR has been proven to be effective in cancer therapy in preclinical and clinical studies. Importantly, tumor-specific alterations such as ATM loss and Cyclin E1 (CCNE1) amplification are more sensitive to ATR inhibition and are being exploited in synthetic lethality (SL) strategy. Besides SL, synergistic anticancer effects involving ATRi have been reported in an increasing number in recent years. This review focuses on the recent advances in different forms of synergistic antitumor effects, summarizes the pharmacological benefits and ongoing clinical trials behind the biological mechanism, and provides perspectives for future challenges and opportunities. The hope is to draw awareness to the community that targeting ATR should have great potential in developing effective anticancer medicines.
Collapse
Affiliation(s)
- Li-Wei Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Songwei Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Ying-Hui Yuan
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Jilong Duan
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Nian-Dong Mao
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Zi Hui
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Renren Bai
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
- Correspondence: (R.B.); (T.X.); (X.-Y.Y.); Tel.: +86-571-28860236 (X.-Y.Y.)
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
- Correspondence: (R.B.); (T.X.); (X.-Y.Y.); Tel.: +86-571-28860236 (X.-Y.Y.)
| | - Xiang-Yang Ye
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
- Correspondence: (R.B.); (T.X.); (X.-Y.Y.); Tel.: +86-571-28860236 (X.-Y.Y.)
| |
Collapse
|
7
|
ElHarouni D, Berker Y, Peterziel H, Gopisetty A, Turunen L, Kreth S, Stainczyk SA, Oehme I, Pietiäinen V, Jäger N, Witt O, Schlesner M, Oppermann S. iTReX: Interactive exploration of mono- and combination therapy dose response profiling data. Pharmacol Res 2021; 175:105996. [PMID: 34848323 DOI: 10.1016/j.phrs.2021.105996] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/11/2022]
Abstract
High throughput screening methods, measuring the sensitivity and resistance of tumor cells to drug treatments have been rapidly evolving. Not only do these screens allow correlating response profiles to tumor genomic features for developing novel predictors of treatment response, but they can also add evidence for therapy decision making in precision oncology. Recent analysis methods developed for either assessing single agents or combination drug efficacies enable quantification of dose-response curves with restricted symmetric fit settings. Here, we introduce iTReX, a user-friendly and interactive Shiny/R application, for both the analysis of mono- and combination therapy responses. The application features an extended version of the drug sensitivity score (DSS) based on the integral of an advanced five-parameter dose-response curve model and a differential DSS for combination therapy profiling. Additionally, iTReX includes modules that visualize drug target interaction networks and support the detection of matches between top therapy hits and the sample omics features to enable the identification of druggable targets and biomarkers. iTReX enables the analysis of various quantitative drug or therapy response readouts (e.g. luminescence, fluorescence microscopy) and multiple treatment strategies (drug treatments, radiation). Using iTReX we validate a cost-effective drug combination screening approach and reveal the application's ability to identify potential sample-specific biomarkers based on drug target interaction networks. The iTReX web application is accessible at https://itrex.kitz-heidelberg.de.
Collapse
Affiliation(s)
- Dina ElHarouni
- Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany; Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Yannick Berker
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Heike Peterziel
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Apurva Gopisetty
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Laura Turunen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Sina Kreth
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Sabine A Stainczyk
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Ina Oehme
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Vilja Pietiäinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Natalie Jäger
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Olaf Witt
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany; Department of Pediatric Oncology, Hematology, Immunology and Pulmonology Heidelberg University Hospital, Heidelberg, Germany
| | - Matthias Schlesner
- Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany; Biomedical Informatics, Data Mining and Data Analytics, Faculty of Applied Computer Science and Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Sina Oppermann
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
8
|
Jia R, Kutzner L, Koren A, Runggatscher K, Májek P, Müller AC, Schuster M, Bock C, Loizou JI, Kubicek S, Kralovics R. High-throughput drug screening identifies the ATR-CHK1 pathway as a therapeutic vulnerability of CALR mutated hematopoietic cells. Blood Cancer J 2021; 11:137. [PMID: 34333533 PMCID: PMC8325683 DOI: 10.1038/s41408-021-00531-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Mutations of calreticulin (CALR) are the second most prevalent driver mutations in essential thrombocythemia and primary myelofibrosis. To identify potential targeted therapies for CALR mutated myeloproliferative neoplasms, we searched for small molecules that selectively inhibit the growth of CALR mutated cells using high-throughput drug screening. We investigated 89 172 compounds using isogenic cell lines carrying CALR mutations and identified synthetic lethality with compounds targeting the ATR-CHK1 pathway. The selective inhibitory effect of these compounds was validated in a co-culture assay of CALR mutated and wild-type cells. Of the tested compounds, CHK1 inhibitors potently depleted CALR mutated cells, allowing wild-type cell dominance in the co-culture over time. Neither CALR deficient cells nor JAK2V617F mutated cells showed hypersensitivity to ATR-CHK1 inhibition, thus suggesting specificity for the oncogenic activation by the mutant CALR. CHK1 inhibitors induced replication stress in CALR mutated cells revealed by elevated pan-nuclear staining for γH2AX and hyperphosphorylation of RPA2. This was accompanied by S-phase cell cycle arrest due to incomplete DNA replication. Transcriptomic and phosphoproteomic analyses revealed a replication stress signature caused by oncogenic CALR, suggesting an intrinsic vulnerability to CHK1 perturbation. This study reveals the ATR-CHK1 pathway as a potential therapeutic target in CALR mutated hematopoietic cells.
Collapse
Affiliation(s)
- Ruochen Jia
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Leon Kutzner
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Anna Koren
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Kathrin Runggatscher
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Peter Májek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - André C Müller
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Michael Schuster
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Joanna I Loizou
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Robert Kralovics
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
9
|
Rødland GE, Hauge S, Hasvold G, Bay LTE, Raabe TTH, Joel M, Syljuåsen RG. Differential Effects of Combined ATR/WEE1 Inhibition in Cancer Cells. Cancers (Basel) 2021; 13:cancers13153790. [PMID: 34359691 PMCID: PMC8345075 DOI: 10.3390/cancers13153790] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 07/13/2021] [Indexed: 01/14/2023] Open
Abstract
Simple Summary Cancer cells often show elevated replication stress and loss of cell cycle checkpoints. The ataxia telangiectasia and Rad3-related (ATR) and WEE1 kinases play roles in protecting cancer cells from high replication stress and in regulating the remaining cell cycle checkpoints. Inhibitors of ATR or WEE1 therefore have the potential to selectively kill cancer cells and are currently being tested in clinical trials. However, more studies are needed to understand how these inhibitors work in various types of cancer and to find the most effective ways of using them. Here, we have explored whether simultaneous treatment with ATR and WEE1 inhibitors is a promising approach. Effects were investigated in cell lines from osteosarcoma and lung cancer. We expect our results to be of importance for future treatment strategies with these inhibitors. Abstract Inhibitors of WEE1 and ATR kinases are considered promising for cancer treatment, either as monotherapy or in combination with chemo- or radiotherapy. Here, we addressed whether simultaneous inhibition of WEE1 and ATR might be advantageous. Effects of the WEE1 inhibitor MK1775 and ATR inhibitor VE822 were investigated in U2OS osteosarcoma cells and in four lung cancer cell lines, H460, A549, H1975, and SW900, with different sensitivities to the WEE1 inhibitor. Despite the differences in cytotoxic effects, the WEE1 inhibitor reduced the inhibitory phosphorylation of CDK, leading to increased CDK activity accompanied by ATR activation in all cell lines. However, combining ATR inhibition with WEE1 inhibition could not fully compensate for cell resistance to the WEE1 inhibitor and reduced cell viability to a variable extent. The decreased cell viability upon the combined treatment correlated with a synergistic induction of DNA damage in S-phase in U2OS cells but not in the lung cancer cells. Moreover, less synergy was found between ATR and WEE1 inhibitors upon co-treatment with radiation, suggesting that single inhibitors may be preferable together with radiotherapy. Altogether, our results support that combining WEE1 and ATR inhibitors may be beneficial for cancer treatment in some cases, but also highlight that the effects vary between cancer cell lines.
Collapse
|
10
|
Branigan TB, Kozono D, Schade AE, Deraska P, Rivas HG, Sambel L, Reavis HD, Shapiro GI, D'Andrea AD, DeCaprio JA. MMB-FOXM1-driven premature mitosis is required for CHK1 inhibitor sensitivity. Cell Rep 2021; 34:108808. [PMID: 33657372 PMCID: PMC7970065 DOI: 10.1016/j.celrep.2021.108808] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/24/2020] [Accepted: 02/09/2021] [Indexed: 12/25/2022] Open
Abstract
To identify genes whose loss confers resistance to CHK1 inhibitors, we perform genome-wide CRISPR-Cas9 screens in non-small-cell lung cancer (NSCLC) cell lines treated with the CHK1 inhibitor prexasertib (CHK1i). Five of the top six hits of the screens, MYBL2 (B-MYB), LIN54, FOXM1, cyclin A2 (CCNA2), and CDC25B, are cell-cycle-regulated genes that contribute to entry into mitosis. Knockout of MMB-FOXM1 complex components LIN54 and FOXM1 reduce CHK1i-induced DNA replication stress markers and premature mitosis during Late S phase. Activation of a feedback loop between the MMB-FOXM1 complex and CDK1 is required for CHK1i-induced premature mitosis in Late S phase and subsequent replication catastrophe, indicating that dysregulation of the S to M transition is necessary for CHK1 inhibitor sensitivity. These findings provide mechanistic insights into small molecule inhibitors currently studied in clinical trials and provide rationale for combination therapies. Branigan et al., by using genome-wide CRISPR screens, identify the MMB-FOXM1 complex as being required for CHK1 inhibitor (CHK1i) sensitivity. Their study shows that CHK1i-induced premature activation of the G2/M transcriptional program by this complex triggers a breakdown in the separation of DNA synthesis and mitosis, leading to replication catastrophe.
Collapse
Affiliation(s)
- Timothy B Branigan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Program in Virology, Graduate School of Arts and Sciences, Harvard University, Cambridge, MA, USA
| | - David Kozono
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Amy E Schade
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Program in Virology, Graduate School of Arts and Sciences, Harvard University, Cambridge, MA, USA
| | - Peter Deraska
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Hembly G Rivas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Program in Virology, Graduate School of Arts and Sciences, Harvard University, Cambridge, MA, USA
| | - Larissa Sambel
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Hunter D Reavis
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Geoffrey I Shapiro
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Alan D D'Andrea
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - James A DeCaprio
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Program in Virology, Graduate School of Arts and Sciences, Harvard University, Cambridge, MA, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
11
|
Carter JL, Hege K, Yang J, Kalpage HA, Su Y, Edwards H, Hüttemann M, Taub JW, Ge Y. Targeting multiple signaling pathways: the new approach to acute myeloid leukemia therapy. Signal Transduct Target Ther 2020; 5:288. [PMID: 33335095 PMCID: PMC7746731 DOI: 10.1038/s41392-020-00361-x] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is the most common form of acute leukemia in adults and the second most common form of acute leukemia in children. Despite this, very little improvement in survival rates has been achieved over the past few decades. This is partially due to the heterogeneity of AML and the need for more targeted therapeutics than the traditional cytotoxic chemotherapies that have been a mainstay in therapy for the past 50 years. In the past 20 years, research has been diversifying the approach to treating AML by investigating molecular pathways uniquely relevant to AML cell proliferation and survival. Here we review the development of novel therapeutics in targeting apoptosis, receptor tyrosine kinase (RTK) signaling, hedgehog (HH) pathway, mitochondrial function, DNA repair, and c-Myc signaling. There has been an impressive effort into better understanding the diversity of AML cell characteristics and here we highlight important preclinical studies that have supported therapeutic development and continue to promote new ways to target AML cells. In addition, we describe clinical investigations that have led to FDA approval of new targeted AML therapies and ongoing clinical trials of novel therapies targeting AML survival pathways. We also describe the complexity of targeting leukemia stem cells (LSCs) as an approach to addressing relapse and remission in AML and targetable pathways that are unique to LSC survival. This comprehensive review details what we currently understand about the signaling pathways that support AML cell survival and the exceptional ways in which we disrupt them.
Collapse
Affiliation(s)
- Jenna L Carter
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA.,MD/PhD Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Katie Hege
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jay Yang
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Hasini A Kalpage
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yongwei Su
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.,National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jeffrey W Taub
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA. .,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA. .,Division of Pediatric Hematology/Oncology, Children's Hospital of Michigan, Detroit, MI, USA. .,Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Yubin Ge
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA. .,Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA. .,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
12
|
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: 0.8] [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
|
13
|
Ghelli Luserna di Rorà A, Cerchione C, Martinelli G, Simonetti G. A WEE1 family business: regulation of mitosis, cancer progression, and therapeutic target. J Hematol Oncol 2020; 13:126. [PMID: 32958072 PMCID: PMC7507691 DOI: 10.1186/s13045-020-00959-2] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/02/2020] [Indexed: 01/05/2023] Open
Abstract
The inhibition of the DNA damage response (DDR) pathway in the treatment of cancer has recently gained interest, and different DDR inhibitors have been developed. Among them, the most promising ones target the WEE1 kinase family, which has a crucial role in cell cycle regulation and DNA damage identification and repair in both nonmalignant and cancer cells. This review recapitulates and discusses the most recent findings on the biological function of WEE1/PKMYT1 during the cell cycle and in the DNA damage repair, with a focus on their dual role as tumor suppressors in nonmalignant cells and pseudo-oncogenes in cancer cells. We here report the available data on the molecular and functional alterations of WEE1/PKMYT1 kinases in both hematological and solid tumors. Moreover, we summarize the preclinical information on 36 chemo/radiotherapy agents, and in particular their effect on cell cycle checkpoints and on the cellular WEE1/PKMYT1-dependent response. Finally, this review outlines the most important pre-clinical and clinical data available on the efficacy of WEE1/PKMYT1 inhibitors in monotherapy and in combination with chemo/radiotherapy agents or with other selective inhibitors currently used or under evaluation for the treatment of cancer patients.
Collapse
Affiliation(s)
- Andrea Ghelli Luserna di Rorà
- Biosciences Laboratory (Onco-hematology Unit), Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | - Claudio Cerchione
- Biosciences Laboratory (Onco-hematology Unit), Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | - Giovanni Martinelli
- Biosciences Laboratory (Onco-hematology Unit), Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | - Giorgia Simonetti
- Biosciences Laboratory (Onco-hematology Unit), Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy.
| |
Collapse
|
14
|
Abstract
DNA damage response (DDR) pathway prevents high level endogenous and environmental DNA damage being replicated and passed on to the next generation of cells via an orchestrated and integrated network of cell cycle checkpoint signalling and DNA repair pathways. Depending on the type of damage, and where in the cell cycle it occurs different pathways are involved, with the ATM-CHK2-p53 pathway controlling the G1 checkpoint or ATR-CHK1-Wee1 pathway controlling the S and G2/M checkpoints. Loss of G1 checkpoint control is common in cancer through TP53, ATM mutations, Rb loss or cyclin E overexpression, providing a stronger rationale for targeting the S/G2 checkpoints. This review will focus on the ATM-CHK2-p53-p21 pathway and the ATR-CHK1-WEE1 pathway and ongoing efforts to target these pathways for patient benefit.
Collapse
|
15
|
ATR-CHK1 pathway as a therapeutic target for acute and chronic leukemias. Cancer Treat Rev 2020; 88:102026. [PMID: 32592909 DOI: 10.1016/j.ctrv.2020.102026] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 12/12/2022]
Abstract
Progress in cancer therapy changed the outcome of many patients and moved therapy from chemotherapy agents to targeted drugs. Targeted drugs already changed the clinical practice in treatment of leukemias, such as imatinib (BCR/ABL inhibitor) in chronic myeloid leukemia (CML) and acute lymphoblastic leukemia (ALL), ibrutinib (Bruton's tyrosine kinase inhibitor) in chronic lymphocytic leukemia (CLL), venetoclax (BCL2 inhibitor) in CLL and acute myeloid leukemia (AML) or midostaurin (FLT3 inhibitor) in AML. In this review, we focused on DNA damage response (DDR) inhibition, specifically on inhibition of ATR-CHK1 pathway. Cancer cells harbor often defects in different DDR pathways, which render them vulnerable to DDR inhibition. Some DDR inhibitors showed interesting single-agent activity even in the absence of cytotoxic drug especially in cancers with underlying defects in DDR or DNA replication. Almost no mutations were found in ATR and CHEK1 genes in leukemia patients. Together with the fact that ATR-CHK1 pathway is essential for cell development and survival of leukemia cells, it represents a promising therapeutic target for treatment of leukemia. ATR-CHK1 inhibition showed excellent results in preclinical testing in acute and chronic leukemias. However, results in clinical trials are so far insufficient. Therefore, the ongoing and future clinical trials will decide on the success of ATR/CHK1 inhibitors in clinical practice of leukemia treatment.
Collapse
|
16
|
Kolb EA, Houghton PJ, Kurmasheva RT, Mosse YP, Maris JM, Erickson SW, Guo Y, Teicher BA, Smith MA, Gorlick R. Preclinical evaluation of the combination of AZD1775 and irinotecan against selected pediatric solid tumors: A Pediatric Preclinical Testing Consortium report. Pediatr Blood Cancer 2020; 67:e28098. [PMID: 31975571 PMCID: PMC8752046 DOI: 10.1002/pbc.28098] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 01/13/2023]
Abstract
INTRODUCTION WEE1 is a serine kinase central to the G2 checkpoint. Inhibition of WEE1 can lead to cell death by permitting cell-cycle progression despite unrepaired DNA damage. AZD1775 is a WEE1 inhibitor that is in clinical development for children and adults with cancer. METHODS AZD1775 was tested using a dose of 120 mg/kg administered orally for days 1 to 5. Irinotecan was administered intraperitoneally at a dose of 2.5 mg/kg for days 1 to 5 (one hour after AZD1775 when used in combination). AZD1775 and irinotecan were studied alone and in combination in neuroblastoma (n = 3), osteosarcoma (n = 4), and Wilms tumor (n = 3) xenografts. RESULTS AZD1775 as a single agent showed little activity. Irinotecan induced objective responses in two neuroblastoma lines (PRs), and two Wilms tumor models (CR and PR). The combination of AZD1775 + irinotecan-induced objective responses in two neuroblastoma lines (PR and CR) and all three Wilms tumor lines (CR and 2 PRs). The objective response measure improved compared with single-agent treatment for one neuroblastoma (PR to CR), two osteosarcoma (PD1 to PD2), and one Wilms tumor (PD2 to PR) xenograft lines. Of note, the combination yielded CR (n = 1) and PR (n = 2) in all the Wilms tumor lines. The event-free survival was significantly longer for the combination compared with single-agent irinotecan in all models tested. The magnitude of the increase was greatest in osteosarcoma and Wilms tumor xenografts. CONCLUSIONS AZD1775 potentiates the effects of irinotecan across most of the xenograft lines tested, with effect size appearing to vary across tumor panels.
Collapse
Affiliation(s)
- E. Anders Kolb
- Nemours Center for Cancer and Blood Disorders, Wilmington, Delaware
| | | | | | - Yael P. Mosse
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - John M. Maris
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | - Yuelong Guo
- RTI International, Research Triangle Park, North Carolina
| | | | | | - Richard Gorlick
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
17
|
Gorecki L, Andrs M, Rezacova M, Korabecny J. Discovery of ATR kinase inhibitor berzosertib (VX-970, M6620): Clinical candidate for cancer therapy. Pharmacol Ther 2020; 210:107518. [PMID: 32109490 DOI: 10.1016/j.pharmthera.2020.107518] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/11/2020] [Indexed: 02/07/2023]
Abstract
Chemoresistance, radioresistance, and the challenge of achieving complete resection are major driving forces in the search for more robust and targeted anticancer therapies. Targeting the DNA damage response has recently attracted research interest, as these processes are enhanced in tumour cells. The major replication stress responder is ATM and Rad3-related (ATR) kinase, which is attracting attention worldwide with four drug candidates currently in phase I/II clinical trials. This review addresses a potent and selective small-molecule ATR inhibitor, which is known as VX-970 (also known as berzosertib or M6620), and summarizes the existing preclinical data to provide deep insight regarding its real potential. We also outline the transition from preclinical to clinical studies, as well as its relationships with other clinical candidates (AZD6738, VX-803 [M4344], and BAY1895344). The results suggest that VX-970 is indeed a promising anticancer drug that can be used both as monotherapy and in combination with either chemotherapy or radiotherapy strategies. Based on patient anamnesis and biomarker identification, VX-970 could become a valuable tool for oncologists in the fight against cancer.
Collapse
Affiliation(s)
- Lukas Gorecki
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Martin Andrs
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; Laboratory of Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Martina Rezacova
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Simkova 870, 500 38 Hradec Kralove, Czech Republic
| | - Jan Korabecny
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic.
| |
Collapse
|
18
|
Tibes R, Bogenberger JM. Transcriptional Silencing of MCL-1 Through Cyclin-Dependent Kinase Inhibition in Acute Myeloid Leukemia. Front Oncol 2019; 9:1205. [PMID: 31921615 PMCID: PMC6920180 DOI: 10.3389/fonc.2019.01205] [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: 07/03/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is the most common adult acute leukemia. Survival remains poor, despite decades of scientific advances. Cytotoxic induction chemotherapy regimens are standard-of-care for most patients. Many investigations have highlighted the genomic heterogeneity of AML, and several new targeted therapeutic options have recently been approved. Additional novel therapies are showing promising clinical results and may rapidly transform the therapeutic landscape of AML. Despite the emerging clinical success of B-cell lymphoma (BCL)-2 targeting in AML and a large body of preclinical data supporting myeloid leukemia cell (MCL)-1 as an attractive therapeutic target for AML, MCL-1 targeting remains relatively unexplored, although novel MCL-1 inhibitors are under clinical investigation. Inhibitors of cyclin-dependent kinases (CDKs) involved in the regulation of transcription, CDK9 in particular, are being investigated in AML as a strategy to target MCL-1 indirectly. In this article, we review the basis for CDK inhibition in oncology with a focus on relevant preclinical mechanism-of-action studies of CDK9 inhibitors in the context of their therapeutic potential specifically in AML.
Collapse
Affiliation(s)
- Raoul Tibes
- NYU School of Medicine & Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
| | | |
Collapse
|
19
|
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: 2.5] [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
|
20
|
Houghton PJ, Kurmasheva RT. Challenges and Opportunities for Childhood Cancer Drug Development. Pharmacol Rev 2019; 71:671-697. [PMID: 31558580 PMCID: PMC6768308 DOI: 10.1124/pr.118.016972] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer in children is rare with approximately 15,700 new cases diagnosed in the United States annually. Through use of multimodality therapy (surgery, radiation therapy, and aggressive chemotherapy), 70% of patients will be "cured" of their disease, and 5-year event-free survival exceeds 80%. However, for patients surviving their malignancy, therapy-related long-term adverse effects are severe, with an estimated 50% having chronic life-threatening toxicities related to therapy in their fourth or fifth decade of life. While overall intensive therapy with cytotoxic agents continues to reduce cancer-related mortality, new understanding of the molecular etiology of many childhood cancers offers an opportunity to redirect efforts to develop effective, less genotoxic therapeutic options, including agents that target oncogenic drivers directly, and the potential for use of agents that target the tumor microenvironment and immune-directed therapies. However, for many high-risk cancers, significant challenges remain.
Collapse
Affiliation(s)
- Peter J Houghton
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| | - Raushan T Kurmasheva
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| |
Collapse
|
21
|
Combining ERBB family and MET inhibitors is an effective therapeutic strategy in cutaneous malignant melanoma independent of BRAF/NRAS mutation status. Cell Death Dis 2019; 10:663. [PMID: 31506424 PMCID: PMC6737096 DOI: 10.1038/s41419-019-1875-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/02/2019] [Accepted: 08/06/2019] [Indexed: 12/17/2022]
Abstract
Current treatment modalities for disseminated cutaneous malignant melanoma (CMM) improve survival; however, relapses are common. A number of receptor tyrosine kinases (RTKs) including EGFR and MET have been reported to be involved in CMM metastasis and in the development of resistance to therapy, targeting the mitogen-activated protein kinase (MAPK pathway). IHC analysis showed that patients with higher MET protein expression had a significantly shorter overall survival. In addition, silencing of MET caused an upregulation of EGFR and p-AKT, which was abrogated by concomitant silencing of MET and EGFR in CMM cells resistant to MAPK-targeting drugs. We therefore explored novel treatment strategies using clinically approved drugs afatinib (ERBB family inhibitor) and crizotinib (MET inhibitor), to simultaneously block MET and ERBB family RTKs. The effects of the combination were assessed in cell culture and spheroid models using established CMM and patient-derived short-term cell lines, and an in vivo xenograft mouse model. The combination had a synergistic effect, promoting cell death, concomitant with a potent downregulation of migratory and invasive capacity independent of their BRAF/NRAS mutational status. Furthermore, the combination attenuated tumor growth rate, as ascertained by the significant reduction of Ki67 expression and induced DNA damage in vivo. Importantly, this combination therapy had minimal therapy-related toxicity in mice. Lastly, the cell cycle G2 checkpoint kinase WEE1 and the RTK IGF1R, non-canonical targets, were altered upon exposure to the combination. Knockdown of WEE1 abrogated the combination-mediated effects on cell migration and proliferation in BRAF mutant BRAF inhibitor-sensitive cells, whereas WEE1 silencing alone inhibited cell migration in NRAS mutant cells. In summary, our results show that afatinib and crizotinib in combination is a promising alternative targeted therapy option for CMM patients, irrespective of BRAF/NRAS mutational status, as well as for cases where resistance has developed towards BRAF inhibitors.
Collapse
|
22
|
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: 2.0] [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
|
23
|
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: 3.2] [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
|
24
|
Lowery CD, Dowless M, Renschler M, Blosser W, VanWye AB, Stephens JR, Iversen PW, Lin AB, Beckmann RP, Krytska K, Cole KA, Maris JM, Hawkins DS, Rubin BP, Kurmasheva RT, Houghton PJ, Gorlick R, Kolb EA, Kang MH, Reynolds CP, Erickson SW, Teicher BA, Smith MA, Stancato LF. Broad Spectrum Activity of the Checkpoint Kinase 1 Inhibitor Prexasertib as a Single Agent or Chemopotentiator Across a Range of Preclinical Pediatric Tumor Models. Clin Cancer Res 2018; 25:2278-2289. [PMID: 30563935 DOI: 10.1158/1078-0432.ccr-18-2728] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 11/13/2018] [Accepted: 12/14/2018] [Indexed: 12/21/2022]
Abstract
PURPOSE Checkpoint kinase 1 (CHK1) inhibitors potentiate the DNA-damaging effects of cytotoxic therapies and/or promote elevated levels of replication stress, leading to tumor cell death. Prexasertib (LY2606368) is a CHK1 small-molecule inhibitor under clinical evaluation in multiple adult and pediatric cancers. In this study, prexasertib was tested in a large panel of preclinical models of pediatric solid malignancies alone or in combination with chemotherapy. EXPERIMENTAL DESIGN DNA damage and changes in cell signaling following in vitro prexasertib treatment in pediatric sarcoma cell lines were analyzed by Western blot and high content imaging. Antitumor activity of prexasertib as a single agent or in combination with different chemotherapies was explored in cell line-derived (CDX) and patient-derived xenograft (PDX) mouse models representing nine different pediatric cancer histologies. RESULTS Pediatric sarcoma cell lines were highly sensitive to prexasertib treatment in vitro, resulting in activation of the DNA damage response. Two PDX models of desmoplastic small round cell tumor and one malignant rhabdoid tumor CDX model responded to prexasertib with complete regression. Prexasertib monotherapy also elicited robust responses in mouse models of rhabdomyosarcoma. Concurrent administration with chemotherapy was sufficient to overcome innate resistance or prevent acquired resistance to prexasertib in preclinical models of neuroblastoma, osteosarcoma, and Ewing sarcoma, or alveolar rhabdomyosarcoma, respectively. CONCLUSIONS Prexasertib has significant antitumor effects as a monotherapy or in combination with chemotherapy in multiple preclinical models of pediatric cancer. These findings support further investigation of prexasertib in pediatric malignancies.
Collapse
Affiliation(s)
- Caitlin D Lowery
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Michele Dowless
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Matthew Renschler
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Wayne Blosser
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Alle B VanWye
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | | | - Philip W Iversen
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Aimee Bence Lin
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | | | - Kateryna Krytska
- The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Kristina A Cole
- The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - John M Maris
- The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Douglas S Hawkins
- Division of Hematology/Oncology, Seattle Children's Hospital, Seattle, Washington
| | - Brian P Rubin
- Departments of Pathology and Cancer Biology, Robert J Tomsich Pathology and Laboratory Medicine Institute and Cleveland Clinic, Cleveland, Ohio
| | | | - Peter J Houghton
- Greehey Children's Cancer Research Institute, San Antonio, Texas
| | | | - E Anders Kolb
- Nemours Center for Cancer and Blood Disorders, Wilmington, Delaware
| | - Min H Kang
- Texas Tech University Health Sciences Center, Lubbock, Texas
| | | | | | | | | | - Louis F Stancato
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana.
| |
Collapse
|
25
|
Chamoun K, Borthakur G. Investigational CHK1 inhibitors in early stage clinical trials for acute myeloid leukemia. Expert Opin Investig Drugs 2018; 27:661-666. [PMID: 30084282 DOI: 10.1080/13543784.2018.1508448] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Introduction: Acute myeloid leukemia (AML) is the most common myeloid malignancy in adults. Despite recent discoveries of targeted therapies, the frontline therapy consisting of chemotherapy remains unchanged for the past four decades. Like other cancers, AML is characterized by deranged DNA damage repair (DDR) pathway. Although impaired DDR may contribute to the pathogenesis of AML it also allows leukemia cells with damaged DNA to attempt repair resulting in resistance. CHK1 inhibitors reverse the cell cycle arrest, disallowing the cell to repair the chemotherapy-induced DNA damage, driving the cell to enter into mitotic catastrophe.Areas covered: This paper reviews the preclinical and clinical development of CHK1 inhibitors and we discussed their promising role as a potential addition to the therapeutic arsenal of AML.Expert opinion: Targeting the cell cycle checkpoints is an intriguing approach to treat cancer in general and AML in particular. CHK1 inhibitors in combination with chemotherapy have the potential of improving outcome in high-risk AML characterized by DDR activation.
Collapse
Affiliation(s)
- Kamal Chamoun
- Leukemia Department, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gautam Borthakur
- Leukemia Department, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
26
|
Duan X, Mu M, Yan J, Bai L, Zhong L, Zhu Y, Pan H, Zhang M, Shi J. Co-delivery of Aurora-A inhibitor XY-4 and Bcl-xl siRNA enhances antitumor efficacy for melanoma therapy. Int J Nanomedicine 2018; 13:1443-1456. [PMID: 29563798 PMCID: PMC5849942 DOI: 10.2147/ijn.s147759] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background The newly synthesized Aurora-A kinase inhibitor XY-4 is a potential anti-cancer agent, but its hydrophobicity and limited efficiency restrict further application. Nanotechnology based combined therapy provides an optimized strategy for solving these issues. Methods In this study, the newly synthesized Aurora-A kinase inhibitor XY-4 and Bcl-xl targeted siRNA were co-delivered by cationic liposomes, creating an injectable co-delivery formulation. The anti-cancer ability and mechanisms of XY-4/Bcl-xl siRNA co-loaded cationic liposomes were studied both in vitro and in vivo. Results The prepared liposomes had a mean particle size of 91.3±4.5 nm with a zeta potential of 38.5±0.5 mV and were monodispersed (Polydispersity index =0.183) in water solution, with high drug loading capacity and stability. Intriguingly, the positive charges of co-delivery liposomes not only facilitated gene delivery, but also obviously enhanced drug uptake. The XY-4/Bcl-xl siRNA co-loaded cationic liposomes demonstrated enhanced anti-cancer effects on B16 melanoma cells in vitro by activation mitochondrial apoptosis pathway. Moreover, intratumoral injection of this co-delivery formulation efficiently inhibited the growth of a B16 melanoma xenograft model in vivo. Conclusion By co-delivering Aurora-A kinase inhibitor XY-4 and Bcl-xl targeting siRNA in a nanoformulation, our study supplied a potential combination strategy for melanoma therapy.
Collapse
Affiliation(s)
- Xingmei Duan
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu
| | - Minjie Mu
- Key Laboratory Standardization of Chinese Herbal Medicines of Ministry of Education, State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Medicine (TCM), Chengdu, People's Republic of China
| | - Junfeng Yan
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu
| | - Lan Bai
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu
| | - Lei Zhong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu
| | - Yuxuan Zhu
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu
| | - Haixia Pan
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu
| | - Mei Zhang
- Key Laboratory Standardization of Chinese Herbal Medicines of Ministry of Education, State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Medicine (TCM), Chengdu, People's Republic of China
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu.,Key Laboratory Standardization of Chinese Herbal Medicines of Ministry of Education, State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Medicine (TCM), Chengdu, People's Republic of China
| |
Collapse
|
27
|
Kim HY, Cho Y, Kang H, Yim YS, Kim SJ, Song J, Chun KH. Targeting the WEE1 kinase as a molecular targeted therapy for gastric cancer. Oncotarget 2018; 7:49902-49916. [PMID: 27363019 PMCID: PMC5226556 DOI: 10.18632/oncotarget.10231] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 05/28/2016] [Indexed: 12/23/2022] Open
Abstract
Wee1 is a member of the Serine/Threonine protein kinase family and is a key regulator of cell cycle progression. It has been known that WEE1 is highly expressed and has oncogenic functions in various cancers, but it is not yet studied in gastric cancers. In this study, we investigated the oncogenic role and therapeutic potency of targeting WEE1 in gastric cancer. At first, higher expression levels of WEE1 with lower survival probability were determined in stage 4 gastric cancer patients or male patients with accompanied lymph node metastasis. To determine the function of WEE1 in gastric cancer cells, we determined that WEE1 ablation decreased the proliferation, migration, and invasion, while overexpression of WEE1 increased these effects in gastric cancer cells. We also validated the clinical application of WEE1 targeting by a small molecule, AZD1775 (MK-1775), which is a WEE1 specific inhibitor undergoing clinical trials. AZD1775 significantly inhibited cell proliferation and induced apoptosis and cell cycle arrest in gastric cancer cells, which was more effective in WEE1 high-expressing gastric cancer cells. Moreover, we performed combination treatments with AZD1775 and anti-cancer agents, 5- fluorouracil or Paclitaxel in gastric cancer cells and in gastric cancer orthotopic-transplanted mice to maximize the therapeutic effect and safety of AZD1775. The combination treatments dramatically inhibited the proliferation of gastric cancer cells and tumor burdens in stomach orthotopic-transplanted mice. Taken together, we propose that WEE1 is over-expressed and could enhance gastric cancer cell proliferation and metastasis. Therefore, we suggest that WEE1 is a potent target for gastric cancer therapy.
Collapse
Affiliation(s)
- Hye-Young Kim
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Seodaemun-gu, Seoul 03722, Korea.,Department of Biochemistry, College of Life Science and Biotechnology, Seodaemun-gu, Seoul 03722, Korea
| | - Yunhee Cho
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Seodaemun-gu, Seoul 03722, Korea.,Brain Korea 21 PlusProject for Medical Science, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - HyeokGu Kang
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Seodaemun-gu, Seoul 03722, Korea.,Brain Korea 21 PlusProject for Medical Science, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Ye-Seal Yim
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Seodaemun-gu, Seoul 03722, Korea.,Brain Korea 21 PlusProject for Medical Science, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Seok-Jun Kim
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Seodaemun-gu, Seoul 03722, Korea.,Brain Korea 21 PlusProject for Medical Science, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Jaewhan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Seodaemun-gu, Seoul 03722, Korea
| | - Kyung-Hee Chun
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Seodaemun-gu, Seoul 03722, Korea.,Brain Korea 21 PlusProject for Medical Science, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| |
Collapse
|
28
|
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: 4.0] [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
|
29
|
Najm FJ, Strand C, Donovan KF, Hegde M, Sanson KR, Vaimberg EW, Sullender ME, Hartenian E, Kalani Z, Fusi N, Listgarten J, Younger ST, Bernstein BE, Root DE, Doench JG. Orthologous CRISPR-Cas9 enzymes for combinatorial genetic screens. Nat Biotechnol 2018; 36:179-189. [PMID: 29251726 PMCID: PMC5800952 DOI: 10.1038/nbt.4048] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 11/04/2017] [Indexed: 12/21/2022]
Abstract
Combinatorial genetic screening using CRISPR-Cas9 is a useful approach to uncover redundant genes and to explore complex gene networks. However, current methods suffer from interference between the single-guide RNAs (sgRNAs) and from limited gene targeting activity. To increase the efficiency of combinatorial screening, we employ orthogonal Cas9 enzymes from Staphylococcus aureus and Streptococcus pyogenes. We used machine learning to establish S. aureus Cas9 sgRNA design rules and paired S. aureus Cas9 with S. pyogenes Cas9 to achieve dual targeting in a high fraction of cells. We also developed a lentiviral vector and cloning strategy to generate high-complexity pooled dual-knockout libraries to identify synthetic lethal and buffering gene pairs across multiple cell types, including MAPK pathway genes and apoptotic genes. Our orthologous approach also enabled a screen combining gene knockouts with transcriptional activation, which revealed genetic interactions with TP53. The "Big Papi" (paired aureus and pyogenes for interactions) approach described here will be widely applicable for the study of combinatorial phenotypes.
Collapse
Affiliation(s)
- Fadi J Najm
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Christine Strand
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | | | - Mudra Hegde
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Kendall R Sanson
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Emma W Vaimberg
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | | | - Ella Hartenian
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Zohra Kalani
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Nicolo Fusi
- Microsoft Research New England, Cambridge, Massachusetts, USA
| | | | - Scott T Younger
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Bradley E Bernstein
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - David E Root
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - John G Doench
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| |
Collapse
|
30
|
Brandsma I, Fleuren ED, Williamson CT, Lord CJ. Directing the use of DDR kinase inhibitors in cancer treatment. Expert Opin Investig Drugs 2017; 26:1341-1355. [PMID: 28984489 PMCID: PMC6157710 DOI: 10.1080/13543784.2017.1389895] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Defects in the DNA damage response (DDR) drive the development of cancer by fostering DNA mutation but also provide cancer-specific vulnerabilities that can be exploited therapeutically. The recent approval of three different PARP inhibitors for the treatment of ovarian cancer provides the impetus for further developing targeted inhibitors of many of the kinases involved in the DDR, including inhibitors of ATR, ATM, CHEK1, CHEK2, DNAPK and WEE1. Areas covered: We summarise the current stage of development of these novel DDR kinase inhibitors, and describe which predictive biomarkers might be exploited to direct their clinical use. Expert opinion: Novel DDR inhibitors present promising candidates in cancer treatment and have the potential to elicit synthetic lethal effects. In order to fully exploit their potential and maximize their utility, identifying highly penetrant predictive biomarkers of single agent and combinatorial DDR inhibitor sensitivity are critical. Identifying the optimal drug combination regimens that could used with DDR inhibitors is also a key objective.
Collapse
Affiliation(s)
- Inger Brandsma
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Emmy D.G. Fleuren
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Chris T. Williamson
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Christopher J. Lord
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| |
Collapse
|
31
|
Mills CC, Kolb EA, Sampson VB. Recent Advances of Cell-Cycle Inhibitor Therapies for Pediatric Cancer. Cancer Res 2017; 77:6489-6498. [PMID: 29097609 DOI: 10.1158/0008-5472.can-17-2066] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/24/2017] [Accepted: 09/19/2017] [Indexed: 12/21/2022]
Abstract
This review describes the pivotal roles of cell-cycle and checkpoint regulators and discusses development of specific cell-cycle inhibitors for therapeutic use for pediatric cancer. The mechanism of action as well as the safety and tolerability of drugs in pediatric patients, including compounds that target CDK4/CDK6 (palbociclib, ribociclib, and abemaciclib), aurora kinases (AT9283 and MLN8237), Wee1 kinase (MK-1775), KSP (ispinesib), and tubulin (taxanes, vinca alkaloids), are presented. The design of mechanism-based combinations that exploit the cross-talk of signals activated by cell-cycle arrest, as well as pediatric-focused drug development, are critical for the advancement of drugs for rare childhood diseases. Cancer Res; 77(23); 6489-98. ©2017 AACR.
Collapse
Affiliation(s)
| | - E A Kolb
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Valerie B Sampson
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware.
| |
Collapse
|
32
|
Randomized phase II trial of cytosine arabinoside with and without the CHK1 inhibitor MK-8776 in relapsed and refractory acute myeloid leukemia. Leuk Res 2017; 61:108-116. [PMID: 28957699 DOI: 10.1016/j.leukres.2017.09.005] [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: 07/18/2017] [Revised: 09/08/2017] [Accepted: 09/10/2017] [Indexed: 11/23/2022]
Abstract
PURPOSE Cytosine arabinoside (AraC) remains the backbone of most treatment regimens for acute myeloid leukemia (AML). Incorporation of AraC into DNA activates checkpoint kinase 1 (Chk1), leading to cell-cycle arrest and diminished AraC cytotoxicity, which can be reversed by the selective Chk1 inhibitor MK-8776. Building on a Phase I trial, we conducted a phase II trial comparing timed sequential AraC with or without MK-8776. METHODS Patients with relapsed or primary refractory AML were randomized 1:1 to receive either AraC with MK-8776 (Arm A); or AraC alone (Arm B). RESULTS 32 patients were treated: 14 assigned to Arm A and 18 to Arm B. There were 5 (36%) complete responses (CR/CRi) and 1 (7%) partial response (PR) in Arm A, and 8 (44%) CR/CRis and 1 (6%) PR in Arm B. Median survival did not differ significantly between the two groups (5.9months in Arm A vs. 4.5 months in Arm B). MK-8776 led to a robust increase in DNA damage in circulating leukemic blasts as measured by increased γ-H2AX (16.9%±6.1% prior and 36.4%±6.8% at one hour after MK-8776 infusion, p=0.016). CONCLUSION Response rates and survival were similar between the two groups in spite of evidence that MK-8776 augmented DNA damage in circulating leukemic blasts. Better than expected results in the control arm using timed sequential AraC and truncated patient enrollment may have limited the ability to detect clinical benefit from the combination.
Collapse
|
33
|
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: 14.1] [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
|
34
|
Suppression of Sirt1 sensitizes lung cancer cells to WEE1 inhibitor MK-1775-induced DNA damage and apoptosis. Oncogene 2017; 36:6863-6872. [DOI: 10.1038/onc.2017.297] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/19/2017] [Accepted: 07/20/2017] [Indexed: 12/16/2022]
|
35
|
Ertz-Archambault N, Kosiorek H, Taylor GE, Kelemen K, Dueck A, Castro J, Marino R, Gauthier S, Finn L, Sproat LZ, Palmer J, Mesa RA, Al-Kali A, Foran J, Tibes R. Association of Therapy for Autoimmune Disease With Myelodysplastic Syndromes and Acute Myeloid Leukemia. JAMA Oncol 2017; 3:936-943. [PMID: 28152123 DOI: 10.1001/jamaoncol.2016.6435] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Importance Therapy-related myeloid neoplasms are a potentially life-threatening consequence of treatment for autoimmune disease (AID) and an emerging clinical phenomenon. Objective To query the association of cytotoxic, anti-inflammatory, and immunomodulating agents to treat patients with AID with the risk for developing myeloid neoplasm. Design, Setting, and Participants This retrospective case-control study and medical record review included 40 011 patients with an International Classification of Diseases, Ninth Revision, coded diagnosis of primary AID who were seen at 2 centers from January 1, 2004, to December 31, 2014; of these, 311 patients had a concomitant coded diagnosis of myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML). Eighty-six cases met strict inclusion criteria. A case-control match was performed at a 2:1 ratio. Main Outcomes and Measures Odds ratio (OR) assessment for AID-directed therapies. Results Among the 86 patients who met inclusion criteria (49 men [57%]; 37 women [43%]; mean [SD] age, 72.3 [15.6] years), 55 (64.0%) had MDS, 21 (24.4%) had de novo AML, and 10 (11.6%) had AML and a history of MDS. Rheumatoid arthritis (23 [26.7%]), psoriasis (18 [20.9%]), and systemic lupus erythematosus (12 [14.0%]) were the most common autoimmune profiles. Median time from onset of AID to diagnosis of myeloid neoplasm was 8 (interquartile range, 4-15) years. A total of 57 of 86 cases (66.3%) received a cytotoxic or an immunomodulating agent. In the comparison group of 172 controls (98 men [57.0%]; 74 women [43.0%]; mean [SD] age, 72.7 [13.8] years), 105 (61.0%) received either agent (P = .50). Azathioprine sodium use was observed more frequently in cases (odds ratio [OR], 7.05; 95% CI, 2.35- 21.13; P < .001). Notable but insignificant case cohort use among cytotoxic agents was found for exposure to cyclophosphamide (OR, 3.58; 95% CI, 0.91-14.11) followed by mitoxantrone hydrochloride (OR, 2.73; 95% CI, 0.23-33.0). Methotrexate sodium (OR, 0.60; 95% CI, 0.29-1.22), mercaptopurine (OR, 0.62; 95% CI, 0.15-2.53), and mycophenolate mofetil hydrochloride (OR, 0.66; 95% CI, 0.21-2.03) had favorable ORs that were not statistically significant. No significant association between a specific length of time of exposure to an agent and the drug's category was observed. Conclusions and Relevance In a large population with primary AID, azathioprine exposure was associated with a 7-fold risk for myeloid neoplasm. The control and case cohorts had similar systemic exposures by agent category. No association was found for anti-tumor necrosis factor agents. Finally, no timeline was found for the association of drug exposure with the incidence in development of myeloid neoplasm.
Collapse
Affiliation(s)
| | - Heidi Kosiorek
- Division of Health Sciences Research, Section of Biostatistics, Mayo Clinic, Phoenix, Arizona
| | | | - Katalin Kelemen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, Arizona
| | - Amylou Dueck
- Division of Health Sciences Research, Section of Biostatistics, Mayo Clinic, Phoenix, Arizona
| | - Janna Castro
- Department of Information Technology, Mayo Clinic, Phoenix, Arizona
| | - Robert Marino
- Division of Planning and Practice Analysis, Mayo Clinic, Phoenix, Arizona
| | - Susanne Gauthier
- Division of Planning and Practice Analysis, Mayo Clinic, Phoenix, Arizona
| | - Laura Finn
- Department of Hematology and Oncology, Mayo Clinic, Rochester, Minnesota
| | - Lisa Z Sproat
- Department of Hematology and Oncology, Mayo Clinic, Phoenix, Arizona
| | - Jeanne Palmer
- Department of Hematology and Oncology, Mayo Clinic, Phoenix, Arizona
| | - Ruben A Mesa
- Department of Hematology and Oncology, Mayo Clinic, Phoenix, Arizona
| | - Aref Al-Kali
- Department of Hematology and Oncology, Mayo Clinic, Rochester, Minnesota
| | - James Foran
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, Florida
| | - Raoul Tibes
- Department of Hematology and Oncology, Mayo Clinic, Phoenix, Arizona
| |
Collapse
|
36
|
McArthur K, D'Cruz AA, Segal D, Lackovic K, Wilks AF, O'Donnell JA, Nowell CJ, Gerlic M, Huang DCS, Burns CJ, Croker BA. Defining a therapeutic window for kinase inhibitors in leukemia to avoid neutropenia. Oncotarget 2017; 8:57948-57963. [PMID: 28938529 PMCID: PMC5601625 DOI: 10.18632/oncotarget.19678] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 06/09/2017] [Indexed: 11/25/2022] Open
Abstract
Neutropenia represents one of the major dose-limiting toxicities of many current cancer therapies. To circumvent the off-target effects of cytotoxic chemotherapeutics, kinase inhibitors are increasingly being used as an adjunct therapy to target leukemia. In this study, we conducted a screen of leukemic cell lines in parallel with primary neutrophils to identify kinase inhibitors with the capacity to induce apoptosis of myeloid and lymphoid cell lines whilst sparing primary mouse and human neutrophils. We have utilized a high-throughput live cell imaging platform to demonstrate that cytotoxic drugs have limited effects on neutrophil viability but are toxic to hematopoietic progenitor cells, with the exception of the topoisomerase I inhibitor SN-38. The parallel screening of kinase inhibitors revealed that mouse and human neutrophil viability is dependent on cyclin-dependent kinase (CDK) activity but surprisingly only partially dependent on PI3 kinase and JAK/STAT signaling, revealing dominant pathways contributing to neutrophil viability. Mcl-1 haploinsufficiency sensitized neutrophils to CDK inhibition, demonstrating that Mcl-1 is a direct target for CDK inhibitors. This study reveals a therapeutic window for the kinase inhibitors BEZ235, BMS-3, AZD7762, and (R)-BI-2536 to induce apoptosis of leukemia cell lines whilst maintaining immunocompetence and hemostasis.
Collapse
Affiliation(s)
- Kate McArthur
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Akshay A D'Cruz
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - David Segal
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Kurt Lackovic
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Andrew F Wilks
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Joanne A O'Donnell
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.,Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Cameron J Nowell
- Monash Institute of Pharmaceutical Sciences, Melbourne, VIC, Australia
| | - Motti Gerlic
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel
| | - David C S Huang
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Christopher J Burns
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.,School of Chemistry, Bio21, The University of Melbourne, Melbourne, VIC, Australia
| | - Ben A Croker
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
37
|
Combined inhibition of Wee1 and Chk1 gives synergistic DNA damage in S-phase due to distinct regulation of CDK activity and CDC45 loading. Oncotarget 2017; 8:10966-10979. [PMID: 28030798 PMCID: PMC5355238 DOI: 10.18632/oncotarget.14089] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/15/2016] [Indexed: 12/24/2022] Open
Abstract
Recent studies have shown synergistic cytotoxic effects of simultaneous Chk1- and Wee1-inhibition. However, the mechanisms behind this synergy are not known. Here, we present a flow cytometry-based screen for compounds that cause increased DNA damage in S-phase when combined with the Wee1-inhibitor MK1775. Strikingly, the Chk1-inhibitors AZD7762 and LY2603618 were among the top candidate hits of 1664 tested compounds, suggesting that the synergistic cytotoxic effects are due to increased S-phase DNA damage. Combined Wee1- and Chk1-inhibition caused a strong synergy in induction of S-phase DNA damage and reduction of clonogenic survival. To address the underlying mechanisms, we developed a novel assay measuring CDK-dependent phosphorylations in single S-phase cells. Surprisingly, while Wee1-inhibition alone induced less DNA damage compared to Chk1-inhibition, Wee1-inhibition caused a bigger increase in S-phase CDK-activity. However, the loading of replication initiation factor CDC45 was more increased after Chk1- than Wee1-inhibition and further increased by the combined treatment, and thus correlated well with DNA damage. Therefore, when Wee1 alone is inhibited, Chk1 suppresses CDC45 loading and thereby limits the extent of unscheduled replication initiation and subsequent S-phase DNA damage, despite very high CDK-activity. These results can explain why combined treatment with Wee1- and Chk1-inhibitors gives synergistic anti-cancer effects.
Collapse
|
38
|
Samadder P, Suchánková T, Hylse O, Khirsariya P, Nikulenkov F, Drápela S, Straková N, Vaňhara P, Vašíčková K, Kolářová H, Binó L, Bittová M, Ovesná P, Kollár P, Fedr R, Ešner M, Jaroš J, Hampl A, Krejčí L, Paruch K, Souček K. Synthesis and Profiling of a Novel Potent Selective Inhibitor of CHK1 Kinase Possessing Unusual N-trifluoromethylpyrazole Pharmacophore Resistant to Metabolic N-dealkylation. Mol Cancer Ther 2017; 16:1831-1842. [PMID: 28619751 DOI: 10.1158/1535-7163.mct-17-0018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 05/21/2017] [Accepted: 06/08/2017] [Indexed: 11/16/2022]
Abstract
Checkpoint-mediated dependency of tumor cells can be deployed to selectively kill them without substantial toxicity to normal cells. Specifically, loss of CHK1, a serine threonine kinase involved in the surveillance of the G2-M checkpoint in the presence of replication stress inflicted by DNA-damaging drugs, has been reported to dramatically influence the viability of tumor cells. CHK1's pivotal role in maintaining genomic stability offers attractive opportunity for increasing the selectivity, effectivity, and reduced toxicity of chemotherapy. Some recently identified CHK1 inhibitors entered clinical trials in combination with DNA antimetabolites. Herein, we report synthesis and profiling of MU380, a nontrivial analogue of clinically profiled compound SCH900776 possessing the highly unusual N-trifluoromethylpyrazole motif, which was envisioned not to undergo metabolic oxidative dealkylation and thereby provide greater robustness to the compound. MU380 is a selective and potent inhibitor of CHK1 which sensitizes a variety of tumor cell lines to hydroxyurea or gemcitabine up to 10 times. MU380 shows extended inhibitory effects in cells, and unlike SCH900776, does not undergo in vivo N-dealkylation to the significantly less selective metabolite. Compared with SCH900776, MU380 in combination with GEM causes higher accumulation of DNA damage in tumor cells and subsequent enhanced cell death, and is more efficacious in the A2780 xenograft mouse model. Overall, MU380 represents a novel state-of-the-art CHK1 inhibitor with high potency, selectivity, and improved metabolic robustness to oxidative N-dealkylation. Mol Cancer Ther; 16(9); 1831-42. ©2017 AACR.
Collapse
Affiliation(s)
- Pounami Samadder
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,National Centre for Biomolecular Research, Masaryk University, Brno, Czech Republic.,International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czech Republic
| | - Tereza Suchánková
- Department of Cytokinetics, Institute of Biophysics CAS, Brno, Czech Republic
| | - Ondřej Hylse
- Department of Chemistry, CZ Openscreen, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Prashant Khirsariya
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czech Republic.,Department of Chemistry, CZ Openscreen, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Fedor Nikulenkov
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czech Republic
| | - Stanislav Drápela
- Department of Cytokinetics, Institute of Biophysics CAS, Brno, Czech Republic
| | - Nicol Straková
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czech Republic.,Department of Cytokinetics, Institute of Biophysics CAS, Brno, Czech Republic
| | - Petr Vaňhara
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czech Republic.,Institute of Biostatistics and Analyses, Masaryk University, Brno, Czech Republic
| | - Kateřina Vašíčková
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czech Republic.,Institute of Biostatistics and Analyses, Masaryk University, Brno, Czech Republic
| | - Hana Kolářová
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,National Centre for Biomolecular Research, Masaryk University, Brno, Czech Republic.,International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czech Republic
| | - Lucia Binó
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czech Republic.,Department of Cytokinetics, Institute of Biophysics CAS, Brno, Czech Republic
| | - Miroslava Bittová
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czech Republic.,Department of Chemistry, CZ Openscreen, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Petra Ovesná
- Department of Cytokinetics, Institute of Biophysics CAS, Brno, Czech Republic.,Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| | - Peter Kollár
- Cellular Imaging Core Facility - CELLIM, CEITEC Masaryk University, Brno, Czech Republic
| | - Radek Fedr
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czech Republic.,Department of Cytokinetics, Institute of Biophysics CAS, Brno, Czech Republic
| | - Milan Ešner
- Institute of Biostatistics and Analyses, Masaryk University, Brno, Czech Republic.,Cellular Imaging Core Facility - CELLIM, CEITEC Masaryk University, Brno, Czech Republic
| | - Josef Jaroš
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czech Republic.,Institute of Biostatistics and Analyses, Masaryk University, Brno, Czech Republic
| | - Aleš Hampl
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czech Republic.,Institute of Biostatistics and Analyses, Masaryk University, Brno, Czech Republic
| | - Lumír Krejčí
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic. .,National Centre for Biomolecular Research, Masaryk University, Brno, Czech Republic.,International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czech Republic
| | - Kamil Paruch
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czech Republic. .,Department of Chemistry, CZ Openscreen, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Karel Souček
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czech Republic. .,Department of Cytokinetics, Institute of Biophysics CAS, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| |
Collapse
|
39
|
Abstract
INTRODUCTION AML therapy remains very challenging despite our increased understanding of its molecular heterogeneity. Outcomes with chemotherapy and targeted therapy remain poor. Targeting cell cycle regulators might complement chemotherapy and targeted therapy and help in improving outcomes. Areas covered: Here we cover the pre-clinical and clinical data for both for cyclin dependent kinase (CDK) and cell-cycle checkpoint inhibitors. While CDK inhibition can inhibit proliferation, checkpoint inhibitors can facilitate cell cycle progression in presence of DNA damage and can induce mitotic catastrophe. Expert opinion: Though the preclinical data for cell cycle inhibitors in AML is compelling, the clinical translation so far has proven to be challenging. This is a reflection of the complexity of both, AML and cell cycle regulators. However, early introduction of cell-cycle active agents in combination with chemotherapy or targeted agents, identifying right sequence of use and identifying right biomarkers might pave the way into successful clinical translation.
Collapse
Affiliation(s)
- Abdallah Abou Zahr
- a Department of Leukemia , University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Gautam Borthakur
- a Department of Leukemia , University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
40
|
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: 60] [Impact Index Per Article: 7.5] [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
|
41
|
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: 208] [Impact Index Per Article: 23.1] [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
|
42
|
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: 4.0] [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
|
43
|
Qi W, Zhang W, Edwards H, Chu R, Madlambayan GJ, Taub JW, Wang Z, Wang Y, Li C, Lin H, Ge Y. Synergistic anti-leukemic interactions between panobinostat and MK-1775 in acute myeloid leukemia ex vivo. Cancer Biol Ther 2016; 16:1784-93. [PMID: 26529495 DOI: 10.1080/15384047.2015.1095406] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
MK-1775 is the first-in-class selective Wee1 inhibitor which has been demonstrated to synergize with CHK1 inhibitors in various malignancies. In this study, we report that the pan-histone deacetylase inhibitor (HDACI) panobinostat synergizes with MK-1775 in acute myeloid leukemia (AML), a malignancy which remains a clinical challenge and requires more effective therapies. Using both AML cell line models and primary patient samples, we demonstrated that panobinostat and MK-1775 synergistically induced proliferation arrest and cell death. We also demonstrated that panobinostat had equal anti-leukemic activities against primary AML blasts derived from patients either at initial diagnosis or at relapse. Interestingly, treatment with panobinostat alone or in combination with MK-1775 resulted in decreased Wee1 protein levels as well as downregulation of the CHK1 pathway. shRNA knockdown of CHK1 significantly sensitized AML cells to MK-1775 treatment, while knockdown of Wee1 significantly enhanced both MK-1775- and panobinostat-induced cell death. Our results demonstrate that panobinostat synergizes with MK-1775 in AML cells, at least in part through downregulation of CHK1 and/or Wee1, providing compelling evidence for the clinical development of the combination treatment in AML.
Collapse
Affiliation(s)
- Wenxiu Qi
- a National Engineering Laboratory for AIDS Vaccine; Key Laboratory for Molecular Enzymology and Engineering; the Ministry of Education; School of Life Sciences; Jilin University ; Changchun , China
| | - Wenbo Zhang
- a National Engineering Laboratory for AIDS Vaccine; Key Laboratory for Molecular Enzymology and Engineering; the Ministry of Education; School of Life Sciences; Jilin University ; Changchun , China
| | - Holly Edwards
- b Department of Oncology ; Wayne State University School of Medicine ; Detroit , MI USA.,c Molecular Therapeutics Program; Barbara Ann Karmanos Cancer Institute; Wayne State University School of Medicine ; Detroit , MI USA
| | - Roland Chu
- d Department of Pediatrics ; Wayne State University School of Medicine ; Detroit , MI USA.,e Division of Pediatric Hematology/Oncology; Children's Hospital of Michigan ; Detroit , MI USA
| | | | - Jeffrey W Taub
- c Molecular Therapeutics Program; Barbara Ann Karmanos Cancer Institute; Wayne State University School of Medicine ; Detroit , MI USA.,d Department of Pediatrics ; Wayne State University School of Medicine ; Detroit , MI USA.,e Division of Pediatric Hematology/Oncology; Children's Hospital of Michigan ; Detroit , MI USA
| | - Zhihong Wang
- d Department of Pediatrics ; Wayne State University School of Medicine ; Detroit , MI USA.,e Division of Pediatric Hematology/Oncology; Children's Hospital of Michigan ; Detroit , MI USA
| | - Yue Wang
- f Department of Pediatric Hematology and Oncology; The First Hospital of Jilin University ; Cangchun , China
| | - Chunhuai Li
- f Department of Pediatric Hematology and Oncology; The First Hospital of Jilin University ; Cangchun , China
| | - Hai Lin
- g Department of Hematology and Oncology; The First Hospital of Jilin University ; Changchun , China
| | - Yubin Ge
- b Department of Oncology ; Wayne State University School of Medicine ; Detroit , MI USA.,c Molecular Therapeutics Program; Barbara Ann Karmanos Cancer Institute; Wayne State University School of Medicine ; Detroit , MI USA
| |
Collapse
|
44
|
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: 12] [Impact Index Per Article: 1.3] [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
|
45
|
Zhang J, Dai Q, Park D, Deng X. Targeting DNA Replication Stress for Cancer Therapy. Genes (Basel) 2016; 7:genes7080051. [PMID: 27548226 PMCID: PMC4999839 DOI: 10.3390/genes7080051] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/06/2016] [Accepted: 08/15/2016] [Indexed: 01/09/2023] Open
Abstract
The human cellular genome is under constant stress from extrinsic and intrinsic factors, which can lead to DNA damage and defective replication. In normal cells, DNA damage response (DDR) mediated by various checkpoints will either activate the DNA repair system or induce cellular apoptosis/senescence, therefore maintaining overall genomic integrity. Cancer cells, however, due to constitutive growth signaling and defective DDR, may exhibit “replication stress” —a phenomenon unique to cancer cells that is described as the perturbation of error-free DNA replication and slow-down of DNA synthesis. Although replication stress has been proven to induce genomic instability and tumorigenesis, recent studies have counterintuitively shown that enhancing replicative stress through further loosening of the remaining checkpoints in cancer cells to induce their catastrophic failure of proliferation may provide an alternative therapeutic approach. In this review, we discuss the rationale to enhance replicative stress in cancer cells, past approaches using traditional radiation and chemotherapy, and emerging approaches targeting the signaling cascades induced by DNA damage. We also summarize current clinical trials exploring these strategies and propose future research directions including the use of combination therapies, and the identification of potential new targets and biomarkers to track and predict treatment responses to targeting DNA replication stress.
Collapse
Affiliation(s)
- Jun Zhang
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA.
| | - Qun Dai
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA.
| | - Dongkyoo Park
- Division of Cancer Biology, Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, 1365C Clifton Road NE, Atlanta, GA 30322, USA.
| | - Xingming Deng
- Division of Cancer Biology, Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, 1365C Clifton Road NE, Atlanta, GA 30322, USA.
| |
Collapse
|
46
|
Zhou L, Chen S, Zhang Y, Kmieciak M, Leng Y, Li L, Lin H, Rizzo KA, Dumur CI, Ferreira-Gonzalez A, Rahmani M, Povirk L, Chalasani S, Berger AJ, Dai Y, Grant S. The NAE inhibitor pevonedistat interacts with the HDAC inhibitor belinostat to target AML cells by disrupting the DDR. Blood 2016; 127:2219-2230. [PMID: 26851293 PMCID: PMC4859196 DOI: 10.1182/blood-2015-06-653717] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 01/27/2016] [Indexed: 02/05/2023] Open
Abstract
Two classes of novel agents, NEDD8-activating enzyme (NAE) and histone deacetylase (HDAC) inhibitors, have shown single-agent activity in acute myelogenous leukemia (AML)/myelodysplastic syndrome (MDS). Here we examined mechanisms underlying interactions between the NAE inhibitor pevonedistat (MLN4924) and the approved HDAC inhibitor belinostat in AML/MDS cells. MLN4924/belinostat coadministration synergistically induced AML cell apoptosis with or without p53 deficiency or FLT3-internal tandem duplication (ITD), whereas p53 short hairpin RNA (shRNA) knockdown or enforced FLT3-ITD expression significantly sensitized cells to the regimen. MLN4924 blocked belinostat-induced antiapoptotic gene expression through nuclear factor-κB inactivation. Each agent upregulated Bim, and Bim knockdown significantly attenuated apoptosis. Microarrays revealed distinct DNA damage response (DDR) genetic profiles between individual vs combined MLN4924/belinostat exposure. Whereas belinostat abrogated the MLN4924-activated intra-S checkpoint through Chk1 and Wee1 inhibition/downregulation, cotreatment downregulated multiple homologous recombination and nonhomologous end-joining repair proteins, triggering robust double-stranded breaks, chromatin pulverization, and apoptosis. Consistently, Chk1 or Wee1 shRNA knockdown significantly sensitized AML cells to MLN4924. MLN4924/belinostat displayed activity against primary AML or MDS cells, including those carrying next-generation sequencing-defined poor-prognostic cancer hotspot mutations, and CD34(+)/CD38(-)/CD123(+) populations, but not normal CD34(+) progenitors. Finally, combined treatment markedly reduced tumor burden and significantly prolonged animal survival (P < .0001) in AML xenograft models with negligible toxicity, accompanied by pharmacodynamic effects observed in vitro. Collectively, these findings argue that MLN4924 and belinostat interact synergistically by reciprocally disabling the DDR in AML/MDS cells. This strategy warrants further consideration in AML/MDS, particularly in disease with unfavorable genetic aberrations.
Collapse
Affiliation(s)
- Liang Zhou
- Division of Hematology and Oncology, Department of Medicine, Virginia Commonwealth University and the Massey Cancer Center, Richmond, VA
| | - Shuang Chen
- Division of Hematology and Oncology, Department of Medicine, Virginia Commonwealth University and the Massey Cancer Center, Richmond, VA
| | - Yu Zhang
- Division of Hematology and Oncology, Department of Medicine, Virginia Commonwealth University and the Massey Cancer Center, Richmond, VA
| | - Maciej Kmieciak
- Division of Hematology and Oncology, Department of Medicine, Virginia Commonwealth University and the Massey Cancer Center, Richmond, VA
| | - Yun Leng
- Division of Hematology and Oncology, Department of Medicine, Virginia Commonwealth University and the Massey Cancer Center, Richmond, VA; Department of Hematology, Beijing Chaoyang Hospital of Capital Medical University, Beijing, China
| | - Lihong Li
- Division of Hematology and Oncology, Department of Medicine, Virginia Commonwealth University and the Massey Cancer Center, Richmond, VA; Department of Hematology, Beijing Chaoyang Hospital of Capital Medical University, Beijing, China
| | - Hui Lin
- Division of Hematology and Oncology, Department of Medicine, Virginia Commonwealth University and the Massey Cancer Center, Richmond, VA
| | - Kathryn A Rizzo
- Division of Molecular Diagnostics, Department of Pathology and
| | | | | | - Mohamed Rahmani
- Division of Hematology and Oncology, Department of Medicine, Virginia Commonwealth University and the Massey Cancer Center, Richmond, VA
| | - Lawrence Povirk
- Department of Pharmacology, Virginia Commonwealth University and the Massey Cancer Center, Richmond, VA
| | - Sri Chalasani
- Department of Pharmacology, Virginia Commonwealth University and the Massey Cancer Center, Richmond, VA
| | - Allison J Berger
- Oncology Drug Discovery Unit, Millennium Pharmaceuticals, Inc/Takeda Pharmaceutical Company Ltd, Cambridge, MA
| | - Yun Dai
- Division of Hematology and Oncology, Department of Medicine, Virginia Commonwealth University and the Massey Cancer Center, Richmond, VA; Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China; and
| | - Steven Grant
- Division of Hematology and Oncology, Department of Medicine, Virginia Commonwealth University and the Massey Cancer Center, Richmond, VA; Department of Biochemistry, Virginia Commonwealth University and the Massey Cancer Center and the Virginia Institute of Molecular Medicine, Richmond, VA
| |
Collapse
|
47
|
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: 5.4] [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
|
48
|
Samadder P, Aithal R, Belan O, Krejci L. Cancer TARGETases: DSB repair as a pharmacological target. Pharmacol Ther 2016; 161:111-131. [PMID: 26899499 DOI: 10.1016/j.pharmthera.2016.02.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer is a disease attributed to the accumulation of DNA damages due to incapacitation of DNA repair pathways resulting in genomic instability and a mutator phenotype. Among the DNA lesions, double stranded breaks (DSBs) are the most toxic forms of DNA damage which may arise as a result of extrinsic DNA damaging agents or intrinsic replication stress in fast proliferating cancer cells. Accurate repair of DSBs is therefore paramount to the cell survival, and several classes of proteins such as kinases, nucleases, helicases or core recombinational proteins have pre-defined jobs in precise execution of DSB repair pathways. On one hand, the proper functioning of these proteins ensures maintenance of genomic stability in normal cells, and on the other hand results in resistance to various drugs employed in cancer therapy and therefore presents a suitable opportunity for therapeutic targeting. Higher relapse and resistance in cancer patients due to non-specific, cytotoxic therapies is an alarming situation and it is becoming more evident to employ personalized treatment based on the genetic landscape of the cancer cells. For the success of personalized treatment, it is of immense importance to identify more suitable targetable proteins in DSB repair pathways and also to explore new synthetic lethal interactions with these pathways. Here we review the various alternative approaches to target the various protein classes termed as cancer TARGETases in DSB repair pathway to obtain more beneficial and selective therapy.
Collapse
Affiliation(s)
- Pounami Samadder
- National Centre for Biomolecular Research, Masaryk University, 62500 Brno, Czech Republic; International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, 60200 Brno, Czech Republic
| | - Rakesh Aithal
- National Centre for Biomolecular Research, Masaryk University, 62500 Brno, Czech Republic; Department of Biology, Masaryk University, 62500 Brno, Czech Republic
| | - Ondrej Belan
- Department of Biology, Masaryk University, 62500 Brno, Czech Republic
| | - Lumir Krejci
- National Centre for Biomolecular Research, Masaryk University, 62500 Brno, Czech Republic; International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, 60200 Brno, Czech Republic; Department of Biology, Masaryk University, 62500 Brno, Czech Republic.
| |
Collapse
|
49
|
Abstract
Over the past decade, rapid advances in genomics, proteomics and functional genomics technologies that enable in-depth interrogation of cancer genomes and proteomes and high-throughput analysis of gene function have enabled characterization of the kinome 'at large' in human cancers, providing crucial insights into how members of the protein kinase superfamily are dysregulated in malignancy, the context-dependent functional role of specific kinases in cancer and how kinome remodelling modulates sensitivity to anticancer drugs. The power of these complementary approaches, and the insights gained from them, form the basis of this Analysis article.
Collapse
Affiliation(s)
- Emmy D G Fleuren
- Department of Medical Oncology, Radboud University Medical Centre, Geert Grooteplein-Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Luxi Zhang
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
- University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Jianmin Wu
- Cancer Division, Kinghorn Cancer Centre, Garvan Institute of Medical Research, 370 Victoria Street, Sydney, New South Wales 2010, Australia
| | - Roger J Daly
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
50
|
Abstract
There is an extensive and growing body of evidence that DNA replication stress is a major driver in the development and progression of many cancers, and that these cancers rely heavily on replication stress response pathways for their continued proliferation. This raises the possibility that the pathways that ordinarily protect cells from the accumulation of cancer-causing mutations may actually prove to be effective therapeutic targets for a wide range of malignancies. In this review, we explore the mechanisms by which sustained proliferation can lead to replication stress and genome instability, and discuss how the pattern of mutations observed in human cancers is supportive of this oncogene-induced replication stress model. Finally, we go on to consider the implications of replication stress both as a prognostic indicator and, more encouragingly, as a potential target in cancer treatment.
Collapse
Affiliation(s)
- Elaine M Taylor
- Lancaster Medical School, Faculty of Health & Medicine, Lancaster University, Lancaster, LA1 4YG, UK
| | - Howard D Lindsay
- Lancaster Medical School, Faculty of Health & Medicine, Lancaster University, Lancaster, LA1 4YG, UK
| |
Collapse
|