51
|
Lin AB, McNeely SC, Beckmann RP. Achieving Precision Death with Cell-Cycle Inhibitors that Target DNA Replication and Repair. Clin Cancer Res 2017; 23:3232-3240. [PMID: 28331049 DOI: 10.1158/1078-0432.ccr-16-0083] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/29/2016] [Accepted: 03/15/2017] [Indexed: 11/16/2022]
Abstract
All cancers are characterized by defects in the systems that ensure strict control of the cell cycle in normal tissues. The consequent excess tissue growth can be countered by drugs that halt cell division, and, indeed, the majority of chemotherapeutics developed during the last century work by disrupting processes essential for the cell cycle, particularly DNA synthesis, DNA replication, and chromatid segregation. In certain contexts, the efficacy of these classes of drugs can be impressive, but because they indiscriminately block the cell cycle of all actively dividing cells, their side effects severely constrain the dose and duration with which they can be administered, allowing both normal and malignant cells to escape complete growth arrest. Recent progress in understanding how cancers lose control of the cell cycle, coupled with comprehensive genomic profiling of human tumor biopsies, has shown that many cancers have mutations affecting various regulators and checkpoints that impinge on the core cell-cycle machinery. These defects introduce unique vulnerabilities that can be exploited by a next generation of drugs that promise improved therapeutic windows in patients whose tumors bear particular genomic aberrations, permitting increased dose intensity and efficacy. These developments, coupled with the success of new drugs targeting cell-cycle regulators, have led to a resurgence of interest in cell-cycle inhibitors. This review in particular focuses on the newer strategies that may facilitate better therapeutic targeting of drugs that inhibit the various components that safeguard the fidelity of the fundamental processes of DNA replication and repair. Clin Cancer Res; 23(13); 3232-40. ©2017 AACR.
Collapse
Affiliation(s)
- Aimee Bence Lin
- Early Phase Medical-Oncology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Samuel C McNeely
- Oncology Business Unit-Patient Tailoring, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | - Richard P Beckmann
- Oncology Translational Research, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana.
| |
Collapse
|
52
|
Ebeid K, Ho GN, Salem AK. HPLC-UV method for simultaneous determination of MK-1775 and AZD-7762 in both acetonitrile-aqueous solution and mouse plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1044-1045:70-76. [PMID: 28088043 DOI: 10.1016/j.jchromb.2016.12.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/19/2016] [Accepted: 12/16/2016] [Indexed: 11/24/2022]
Abstract
A sensitive and precise method is described for the simultaneous determination of two small molecule kinase inhibitors: MK-1775 (MK) and AZD-7762 (AZD), in acetonitrile (ACN)-aqueous solution and in mouse plasma. A Nova-Pak C18 reversed phase column (3.9mm×150mm, 4μm, 60Å) was utilized in the separation using an isocratic mobile phase of 0.1% v/v triethylamine in phosphate buffer (pH=7.4): acetonitrile (ACN) (60:40, v/v), at a flow rate of 0.8mL/min. Detection wavelength was set at 310nm for both MK and AZD, and 431nm for the internal standard sunitinib (SUN). The developed method was validated following the ICH guidelines and it was shown to be accurate, precise and linear in the range of 41ng/mL to 8333ng/mL for both drugs in the ACN-aqueous solution and from 83ng/mL to 8333ng/mL for both drugs in mouse plasma samples. For the first time, the presented data suggest the suitability of this method for the simultaneous separation and quantification of MK and AZD in both ACN aqueous solution as well as in mouse plasma samples.
Collapse
Affiliation(s)
- Kareem Ebeid
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 51141, USA
| | - Giang N Ho
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 51141, USA
| | - Aliasger K Salem
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 51141, USA.
| |
Collapse
|
53
|
Restelli V, Vagni M, Arribas AJ, Bertoni F, Damia G, Carrassa L. Inhibition of
CHK
1 and
WEE
1 as a new therapeutic approach in diffuse large B cell lymphomas with
MYC
deregulation. Br J Haematol 2016; 181:129-133. [DOI: 10.1111/bjh.14506] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Valentina Restelli
- Laboratory of Molecular Pharmacology Department of Oncology IRCCS – Istituto di Ricerche Farmacologiche “Mario Negri” Milan Italy
| | - Micaela Vagni
- Laboratory of Molecular Pharmacology Department of Oncology IRCCS – Istituto di Ricerche Farmacologiche “Mario Negri” Milan Italy
| | - Alberto J. Arribas
- Lymphoma and Genomics Research Program IOR Institute of Oncology Research Bellinzona Switzerland
| | - Francesco Bertoni
- Lymphoma and Genomics Research Program IOR Institute of Oncology Research Bellinzona Switzerland
- IOSI Oncology Institute of Southern Switzerland Bellinzona Switzerland
| | - Giovanna Damia
- Laboratory of Molecular Pharmacology Department of Oncology IRCCS – Istituto di Ricerche Farmacologiche “Mario Negri” Milan Italy
| | - Laura Carrassa
- Laboratory of Molecular Pharmacology Department of Oncology IRCCS – Istituto di Ricerche Farmacologiche “Mario Negri” Milan Italy
| |
Collapse
|
54
|
Leijen S, van Geel RM, Pavlick AC, Tibes R, Rosen L, Razak ARA, Lam R, Demuth T, Rose S, Lee MA, Freshwater T, Shumway S, Liang LW, Oza AM, Schellens JH, Shapiro GI. Phase I Study Evaluating WEE1 Inhibitor AZD1775 As Monotherapy and in Combination With Gemcitabine, Cisplatin, or Carboplatin in Patients With Advanced Solid Tumors. J Clin Oncol 2016; 34:4371-4380. [PMID: 27601554 PMCID: PMC7845944 DOI: 10.1200/jco.2016.67.5991] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Purpose AZD1775 is a WEE1 kinase inhibitor targeting G2 checkpoint control, preferentially sensitizing TP53-deficient tumor cells to DNA damage. This phase I study evaluated safety, tolerability, pharmacokinetics, and pharmacodynamics of oral AZD1775 as monotherapy or in combination with chemotherapy in patients with refractory solid tumors. Patients and Methods In part 1, patients received a single dose of AZD1775 followed by 14 days of observation. In part 2, patients received AZD1775 as a single dose (part 2A) or as five twice per day doses or two once per day doses (part 2B) in combination with one of the following chemotherapy agents: gemcitabine (1,000 mg/m2), cisplatin (75 mg/m2), or carboplatin (area under the curve, 5 mg/mL⋅min). Skin biopsies were collected for pharmacodynamic assessments. TP53 status was determined retrospectively in archival tumor tissue. Results Two hundred two patients were enrolled onto the study, including nine patients in part 1, 43 in part 2A (including eight rollover patients from part 1), and 158 in part 2B. AZD1775 monotherapy given as single dose was well tolerated, and the maximum-tolerated dose was not reached. In the combination regimens, the most common adverse events consisted of fatigue, nausea and vomiting, diarrhea, and hematologic toxicity. The maximum-tolerated doses and biologically effective doses were established for each combination. Target engagement, as a predefined 50% pCDK1 reduction in surrogate tissue, was observed in combination with cisplatin and carboplatin. Of 176 patients evaluable for efficacy, 94 (53%) had stable disease as best response, and 17 (10%) achieved a partial response. The response rate in TP53-mutated patients (n = 19) was 21% compared with 12% in TP53 wild-type patients (n = 33). Conclusion AZD1775 was safe and tolerable as a single agent and in combination with chemotherapy at doses associated with target engagement.
Collapse
Affiliation(s)
- Suzanne Leijen
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Robin M.J.M. van Geel
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Anna C. Pavlick
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Raoul Tibes
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Lee Rosen
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Albiruni R. Abdul Razak
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Raymond Lam
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Tim Demuth
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Shelonitda Rose
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Mark A. Lee
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Tomoko Freshwater
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Stuart Shumway
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Li Wen Liang
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Amit M. Oza
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Jan H.M. Schellens
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| | - Geoffrey I. Shapiro
- Suzanne Leijen, Robin M.J.M. van Geel, and Jan H.M. Schellens, The Netherlands Cancer Institute, Amsterdam; Jan H.M. Schellens, Utrecht University, Utrecht, the Netherlands; Anna C. Pavlick, New York University Medical Center, New York, NY; Lee Rosen, University of California Los Angeles, Santa Monica, CA; Raymond Lam, Shelonitda Rose, Mark A. Lee, Tomoko Freshwater, and Stuart Shumway, Merck, Kenilworth, NJ; Geoffrey I. Shapiro, Dana-Farber Cancer Institute, Boston, MA; Albiruni R. Abdul Razak and Amit M. Oza, Princess Margaret Hospital, Toronto, Ontario, Canada; Raoul Tibes, University Hospital of Würzburg, Würzburg; Tim Demuth, Sandoz AG, Holzkirchen, Germany; and Li Wen Liang, Merck Sharp & Dohme R&D, Beijing, China
| |
Collapse
|
55
|
Sakurikar N, Thompson R, Montano R, Eastman A. A subset of cancer cell lines is acutely sensitive to the Chk1 inhibitor MK-8776 as monotherapy due to CDK2 activation in S phase. Oncotarget 2016; 7:1380-94. [PMID: 26595527 PMCID: PMC4811467 DOI: 10.18632/oncotarget.6364] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/16/2015] [Indexed: 12/14/2022] Open
Abstract
DNA damage activates Checkpoint kinase 1 (Chk1) to halt cell cycle progression thereby preventing further DNA replication and mitosis until the damage has been repaired. Consequently, Chk1 inhibitors have emerged as promising anticancer therapeutics in combination with DNA damaging drugs, but their single agent activity also provides a novel approach that may be particularly effective in a subset of patients. From analysis of a large panel of cell lines, we demonstrate that 15% are very sensitive to the Chk1 inhibitor MK-8776. Upon inhibition of Chk1, sensitive cells rapidly accumulate DNA double-strand breaks in S phase in a CDK2- and cyclin A-dependent manner. In contrast, resistant cells can continue to grow for at least 7 days despite continued inhibition of Chk1. Resistance can be circumvented by inhibiting Wee1 kinase and thereby directly activating CDK2. Hence, sensitivity to Chk1 inhibition is regulated upstream of CDK2 and correlates with accumulation of CDC25A. We conclude that cells poorly tolerate CDK2 activity in S phase and that a major function of Chk1 is to ensure it remains inactive. Indeed, inhibitors of CDK1 and CDK2 arrest cells in G1 or G2, respectively, but do not prevent progression through S phase demonstrating that neither kinase is required for S phase progression. Inappropriate activation of CDK2 in S phase underlies the sensitivity of a subset of cell lines to Chk1 inhibitors, and this may provide a novel therapeutic opportunity for appropriately stratified patients.
Collapse
Affiliation(s)
- Nandini Sakurikar
- Department of Pharmacology and Toxicology, and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Ruth Thompson
- Department of Pharmacology and Toxicology, and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Ryan Montano
- Department of Pharmacology and Toxicology, and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Alan Eastman
- Department of Pharmacology and Toxicology, and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
56
|
Busch CJ, Kröger MS, Jensen J, Kriegs M, Gatzemeier F, Petersen C, Münscher A, Rothkamm K, Rieckmann T. G2-checkpoint targeting and radiosensitization of HPV/p16-positive HNSCC cells through the inhibition of Chk1 and Wee1. Radiother Oncol 2016; 122:260-266. [PMID: 27939202 DOI: 10.1016/j.radonc.2016.11.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/07/2016] [Accepted: 11/19/2016] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND PURPOSE HPV-positive HNSCC cells are characterized by radiosensitivity, inefficient DNA double-strand break repair and a profound and prolonged arrest in G2. Here we explored the effect of clinically relevant inhibitors of Chk1 and Wee1 to inhibit the radiation-induced G2-arrest in order to achieve further radiosensitization. MATERIAL AND METHODS Assessment of Chk1 activity by Western blot; assessment of cell cycle distribution by propidium iodide staining and flow cytometry; assessment of cell survival by colony formation assay. HPV+ HNSCC cell lines: UD-SCC-2, UM-SCC-47 and UPCI-SCC-154; Chk1 inhibitors: LY2603618, MK8776; Wee1 inhibitor: AZD1775. RESULTS Specific Chk1 inhibitors efficiently abrogated the radiation-induced G2-arrest and caused radiosensitization. Wee-inhibition by AZD1775 resulted in the activation of Chk1. This feedback mechanism is likely to counteract some of the effects of Wee1 inhibition but could be antagonized through the combined inhibition of both kinases. Combined inhibition was effective using profoundly reduced concentrations of both inhibitors and resulted in more efficient radiosensitization of the HPV-positive cell lines compared to p53 proficient normal human fibroblasts. CONCLUSIONS Specific Chk1 inhibitors as well as the combined inhibition of Chk1 and Wee1 radiosensitize HPV-positive HNSCC cells.
Collapse
Affiliation(s)
- Chia-Jung Busch
- Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Germany
| | - Marie Sophie Kröger
- Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Germany; Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Jana Jensen
- Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Germany; Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Malte Kriegs
- Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Fruzsina Gatzemeier
- Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Germany; Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Cordula Petersen
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Adrian Münscher
- Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Germany
| | - Kai Rothkamm
- Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Thorsten Rieckmann
- Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Germany; Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Germany.
| |
Collapse
|
57
|
Ronco C, Martin AR, Demange L, Benhida R. ATM, ATR, CHK1, CHK2 and WEE1 inhibitors in cancer and cancer stem cells. MEDCHEMCOMM 2016; 8:295-319. [PMID: 30108746 DOI: 10.1039/c6md00439c] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/25/2016] [Indexed: 12/15/2022]
Abstract
DNA inevitably undergoes a high number of damages throughout the cell cycle. To preserve the integrity of the genome, cells have developed a complex enzymatic machinery aimed at sensing and repairing DNA lesions, pausing the cell cycle to provide more time to repair, or induce apoptosis if damages are too severe. This so-called DNA-damage response (DDR) is yet considered as a major source of resistance to DNA-damaging treatments in oncology. Recently, it has been hypothesized that cancer stem cells (CSC), a sub-population of cancer cells particularly resistant and with tumour-initiating ability, allow tumour re-growth and cancer relapse. Therefore, DDR appears as a relevant target to sensitize cancer cells and cancer stem cells to classical radio- and chemotherapies as well as to overcome resistances. Moreover, the concept of synthetic lethality could be particularly efficiently exploited in DDR. Five kinases play pivotal roles in the DDR: ATM, ATR, CHK1, CHK2 and WEE1. Herein, we review the drugs targeting these proteins and the inhibitors used in the specific case of CSC. We also suggest molecules that may be of interest for preclinical and clinical researchers studying checkpoint inhibition to sensitize cancer and cancer stem cells to DNA-damaging treatments.
Collapse
Affiliation(s)
- Cyril Ronco
- Université Côte d'Azur , CNRS , Institut de Chimie de Nice , UMR7272 - Parc Valrose , 06108 Nice Cedex 2 , France . ; ; Tel: +33 4 92076143
| | - Anthony R Martin
- Université Côte d'Azur , CNRS , Institut de Chimie de Nice , UMR7272 - Parc Valrose , 06108 Nice Cedex 2 , France . ; ; Tel: +33 4 92076143
| | - Luc Demange
- Université Côte d'Azur , CNRS , Institut de Chimie de Nice , UMR7272 - Parc Valrose , 06108 Nice Cedex 2 , France . ; ; Tel: +33 4 92076143.,Université Paris Descartes , Sorbonne Paris Cité , UFR des Sciences Pharmaceutiques , 4 avenue de l'Observatoire , Paris Fr-75006 , France.,Université Paris Descartes , Sorbonne Paris Cité , UFR Biomédicale des Saints Pères , 45 rue des Saints Pères , France
| | - Rachid Benhida
- Université Côte d'Azur , CNRS , Institut de Chimie de Nice , UMR7272 - Parc Valrose , 06108 Nice Cedex 2 , France . ; ; Tel: +33 4 92076143
| |
Collapse
|
58
|
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: 3.6] [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
|
59
|
Restelli V, Chilà R, Lupi M, Rinaldi A, Kwee I, Bertoni F, Damia G, Carrassa L. Characterization of a mantle cell lymphoma cell line resistant to the Chk1 inhibitor PF-00477736. Oncotarget 2016; 6:37229-40. [PMID: 26439697 PMCID: PMC4741926 DOI: 10.18632/oncotarget.5954] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/17/2015] [Indexed: 12/24/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell lymphoma characterized by the chromosomal translocation t(11;14) that leads to constitutive expression of cyclin D1, a master regulator of the G1-S phase. Chk1 inhibitors have been recently shown to be strongly effective as single agents in MCL. To investigate molecular mechanisms at the basis of Chk1 inhibitor activity, a MCL cell line resistant to the Chk1 inhibitor PF-00477736 (JEKO-1 R) was obtained and characterized. The JEKO-1 R cell line was cross resistant to another Chk1 inhibitor (AZD-7762) and to the Wee1 inhibitor MK-1775. It displayed a shorter doubling time than parental cell line, likely due to a faster S phase. Cyclin D1 expression levels were decreased in resistant cell line and its re-overexpression partially re-established PF-00477736 sensitivity. Gene expression profiling showed an enrichment in gene sets involved in pro-survival pathways in JEKO-1 R. Dasatinib treatment partly restored PF-00477736 sensitivity in resistant cells suggesting that the pharmacological interference of pro-survival pathways can overcome the resistance to Chk1 inhibitors. These data further corroborate the involvement of the t(11;14) in cellular sensitivity to Chk1 inhibitors, fostering the clinical testing of Chk1 inhibitors as single agents in MCL.
Collapse
Affiliation(s)
- Valentina Restelli
- Laboratory of Molecular Pharmacology and Laboratory of Cancer Pharmacology, Department of Oncology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Rosaria Chilà
- Laboratory of Molecular Pharmacology and Laboratory of Cancer Pharmacology, Department of Oncology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Monica Lupi
- Laboratory of Molecular Pharmacology and Laboratory of Cancer Pharmacology, Department of Oncology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Andrea Rinaldi
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland
| | - Ivo Kwee
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland.,Dalle Molle Institute for Artificial Intelligence (IDSIA), Manno, Switzerland.,Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Francesco Bertoni
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland.,Lymphoma Unit, IOSI Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Giovanna Damia
- Laboratory of Molecular Pharmacology and Laboratory of Cancer Pharmacology, Department of Oncology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Laura Carrassa
- Laboratory of Molecular Pharmacology and Laboratory of Cancer Pharmacology, Department of Oncology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| |
Collapse
|
60
|
Chemogenetic profiling identifies RAD17 as synthetically lethal with checkpoint kinase inhibition. Oncotarget 2016; 6:35755-69. [PMID: 26437225 PMCID: PMC4742139 DOI: 10.18632/oncotarget.5928] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 09/14/2015] [Indexed: 01/07/2023] Open
Abstract
Chemical inhibitors of the checkpoint kinases have shown promise in the treatment of cancer, yet their clinical utility may be limited by a lack of molecular biomarkers to identify specific patients most likely to respond to therapy. To this end, we screened 112 known tumor suppressor genes for synthetic lethal interactions with inhibitors of the CHEK1 and CHEK2 checkpoint kinases. We identified eight interactions, including the Replication Factor C (RFC)-related protein RAD17. Clonogenic assays in RAD17 knockdown cell lines identified a substantial shift in sensitivity to checkpoint kinase inhibition (3.5-fold) as compared to RAD17 wild-type. Additional evidence for this interaction was found in a large-scale functional shRNA screen of over 100 genotyped cancer cell lines, in which CHEK1/2 mutant cell lines were unexpectedly sensitive to RAD17 knockdown. This interaction was widely conserved, as we found that RAD17 interacts strongly with checkpoint kinases in the budding yeast Saccharomyces cerevisiae. In the setting of RAD17 knockdown, CHEK1/2 inhibition was found to be synergistic with inhibition of WEE1, another pharmacologically relevant checkpoint kinase. Accumulation of the DNA damage marker γH2AX following chemical inhibition or transient knockdown of CHEK1, CHEK2 or WEE1 was magnified by knockdown of RAD17. Taken together, our data suggest that CHEK1 or WEE1 inhibitors are likely to have greater clinical efficacy in tumors with RAD17 loss-of-function.
Collapse
|
61
|
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: 7.8] [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
|
62
|
Srivas R, Shen JP, Yang CC, Sun SM, Li J, Gross AM, Jensen J, Licon K, Bojorquez-Gomez A, Klepper K, Huang J, Pekin D, Xu JL, Yeerna H, Sivaganesh V, Kollenstart L, van Attikum H, Aza-Blanc P, Sobol RW, Ideker T. A Network of Conserved Synthetic Lethal Interactions for Exploration of Precision Cancer Therapy. Mol Cell 2016; 63:514-25. [PMID: 27453043 DOI: 10.1016/j.molcel.2016.06.022] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 03/03/2016] [Accepted: 06/15/2016] [Indexed: 01/06/2023]
Abstract
An emerging therapeutic strategy for cancer is to induce selective lethality in a tumor by exploiting interactions between its driving mutations and specific drug targets. Here we use a multi-species approach to develop a resource of synthetic lethal interactions relevant to cancer therapy. First, we screen in yeast ∼169,000 potential interactions among orthologs of human tumor suppressor genes (TSG) and genes encoding drug targets across multiple genotoxic environments. Guided by the strongest signal, we evaluate thousands of TSG-drug combinations in HeLa cells, resulting in networks of conserved synthetic lethal interactions. Analysis of these networks reveals that interaction stability across environments and shared gene function increase the likelihood of observing an interaction in human cancer cells. Using these rules, we prioritize ∼10(5) human TSG-drug combinations for future follow-up. We validate interactions based on cell and/or patient survival, including topoisomerases with RAD17 and checkpoint kinases with BLM.
Collapse
Affiliation(s)
- Rohith Srivas
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA; The Cancer Cell Map Initiative
| | - John Paul Shen
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; The Cancer Cell Map Initiative
| | - Chih Cheng Yang
- Functional Genomics Core, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Su Ming Sun
- Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, the Netherlands
| | - Jianfeng Li
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Andrew M Gross
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - James Jensen
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Katherine Licon
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Ana Bojorquez-Gomez
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Kristin Klepper
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Justin Huang
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel Pekin
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jia L Xu
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Huwate Yeerna
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Vignesh Sivaganesh
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Leonie Kollenstart
- Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, the Netherlands
| | - Haico van Attikum
- Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, the Netherlands
| | - Pedro Aza-Blanc
- Functional Genomics Core, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Robert W Sobol
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Trey Ideker
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; The Cancer Cell Map Initiative.
| |
Collapse
|
63
|
Mak JPY, Man WY, Chow JPH, Ma HT, Poon RYC. Pharmacological inactivation of CHK1 and WEE1 induces mitotic catastrophe in nasopharyngeal carcinoma cells. Oncotarget 2016; 6:21074-84. [PMID: 26025928 PMCID: PMC4673251 DOI: 10.18632/oncotarget.4020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/09/2015] [Indexed: 12/23/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a rare but highly invasive cancer. As radiotherapy is the primary treatment for NPC, this offers a rationale to investigate if uncoupling the DNA damage responses can sensitize this cancer type. The G2 DNA damage checkpoint is controlled by a cascade of protein kinases: ATM/ATR, which phosphorylates CHK1/CHK2, which in turn phosphorylates WEE1. A number of small molecule inhibitors have been developed against these kinases as potential therapeutic agents. Here we demonstrated that compare to that in immortalized nasopharyngeal epithelial cells, ATR, CHK1, and WEE1 were overexpressed in NPC cell lines. Inhibitors of these kinases were unable to promote extensive mitotic catastrophe in ionizing radiation-treated NPC cells, indicating that they are not very effective radiosensitizer for this cancer. In the absence of prior irradiation, however, mitotic catastrophe could be induced with inhibitors against CHK1 (AZD7762) or WEE1 (MK-1775). NPC cells were more sensitive to WEE1 inactivation than nasopharyngeal epithelial cells. Targeting CHK1 and WEE1 together induced more extensive mitotic catastrophe than the individual components alone. Taken together, our results show that NPC cells depend on CHK1 and WEE1 activity for growth and that inhibitors of these kinases may serve as potential therapeutics for NPC.
Collapse
Affiliation(s)
- Joyce P Y Mak
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Wing Yu Man
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Jeremy P H Chow
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Hoi Tang Ma
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Randy Y C Poon
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| |
Collapse
|
64
|
Aung KL, Siu LL. Genomically personalized therapy in head and neck cancer. CANCERS OF THE HEAD & NECK 2016; 1:2. [PMID: 31093332 PMCID: PMC6457141 DOI: 10.1186/s41199-016-0004-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/17/2016] [Indexed: 12/13/2022]
Abstract
The current treatment paradigm in head and neck cancer does not adequately address its clinical and biological heterogeneity. Data from genomic profiling studies in head and neck squamous cell carcinoma (HNSCC) have revealed the molecular features that are unique to HNSCC subgroups. This progress in the understanding of HNSCC biology provides an opportunity to develop personalized therapies for patients with distinct molecular subtypes to achieve better clinical outcomes including survival. However there are several well-recognized challenges that need to be overcome before genotype-matched therapies make precision medicine a reality for patients with HNSCC. Selection of appropriate patients for biomarker directed clinical trials based on sound scientific rationale will be critical in making cancer genomics more applicable in this malignancy.
Collapse
Affiliation(s)
- Kyaw L Aung
- Drug Development Program, Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Suite 5-718, Toronto, ON M5G 2M9 Canada
| | - Lillian L Siu
- Drug Development Program, Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Suite 5-718, Toronto, ON M5G 2M9 Canada
| |
Collapse
|
65
|
Egeland EV, Flatmark K, Nesland JM, Flørenes VA, Mælandsmo GM, Boye K. Expression and clinical significance of Wee1 in colorectal cancer. Tumour Biol 2016; 37:12133-12140. [PMID: 27220319 DOI: 10.1007/s13277-016-5081-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 05/15/2016] [Indexed: 12/22/2022] Open
Abstract
Wee1 is a nuclear kinase regulating cell cycle progression, and has emerged as a promising therapeutic target in cancer. Expression of Wee1 has been associated with poor outcome in certain tumor types, but the prognostic impact and clinical significance in colorectal cancer is unknown. The expression of Wee1 was examined by immunohistochemistry in primary colorectal carcinomas from a prospectively collected patient cohort, and associations with clinicopathological parameters and outcome were investigated. Cell culture experiments were performed using the cell lines RKO and SW620, and the relationship with the metastasis-promoting protein S100A4 was investigated. Nuclear expression was detected in 229 of the 258 tumors analyzed (89 %). Wee1 staining was associated with low pT stage, but no other significant associations with demographic or histopathological variables were found. Moderate Wee1 staining intensity was a predictor of favorable metastasis-free and overall survival compared to strong intensity and no or weak staining. The fraction of positive cells was not a prognostic factor in the present cohort. Inhibition of Wee1 expression using siRNA or treatment with the Wee1 inhibitor MK-1775 reduced expression of the metastasis-promoting protein S100A4, but no relationship between Wee1 and S100A4 was found in the patient samples. In conclusion, Wee1 is highly expressed in primary colorectal carcinomas, but few relevant associations with clinicopathological parameters or outcome were found. The lack of clinical significance of Wee1 expression could indicate that other tumor types might be better suited for further development of Wee1 inhibitors.
Collapse
Affiliation(s)
- Eivind Valen Egeland
- Department of Tumor Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, PO Box 4953, Nydalen, NO-0424, Oslo, Norway
| | - Kjersti Flatmark
- Department of Tumor Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, PO Box 4953, Nydalen, NO-0424, Oslo, Norway.,Department of Gastroenterological Surgery, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Medical Faculty, University of Oslo, Oslo, Norway
| | - Jahn M Nesland
- Medical Faculty, University of Oslo, Oslo, Norway.,Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Vivi Ann Flørenes
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Gunhild M Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, PO Box 4953, Nydalen, NO-0424, Oslo, Norway.,Department of Pharmacy, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Kjetil Boye
- Department of Tumor Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, PO Box 4953, Nydalen, NO-0424, Oslo, Norway. .,Department of Oncology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
66
|
Combined inhibition of Chk1 and Wee1 as a new therapeutic strategy for mantle cell lymphoma. Oncotarget 2016; 6:3394-408. [PMID: 25428911 PMCID: PMC4413661 DOI: 10.18632/oncotarget.2583] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 10/09/2014] [Indexed: 12/13/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive, incurable disease, characterized by a deregulated cell cycle. Chk1 and Wee1 are main regulators of cell cycle progression and recent data on solid tumors suggest that simultaneous inhibition of these proteins has a strong synergistic cytotoxic effect. The effects of a Chk1 inhibitor (PF-00477736) and a Wee1 inhibitor (MK-1775) have been herein investigated in a large panel of mature B-cell lymphoma cell lines. We found that MCL cells were the most sensitive to the Chk1 inhibitor PF-00477736 and Wee1 inhibitor MK-1775 as single agents. Possible involvement of the translocation t(11;14) in Chk1 inhibitor sensitivity was hypothesized. The combined inhibition of Chk1 and Wee1 was strongly synergistic in MCL cells, leading to deregulation of the cell cycle, with increased activity of CDK2 and CDK1, and activation of apoptosis. In vivo treatment with the drug combination of mice bearing JeKo-1 xenografts (MCL) had a marked antitumor effect with tumor regressions observed at non-toxic doses (best T/C%=0.54%). Gene expression profiling suggested effect on genes involved in apoptosis. The strong synergism observed by combining Chk1 and Wee1 inhibitors in preclinical models of MCL provides the rationale for testing this combination in the clinical setting.
Collapse
|
67
|
Blanco FF, Pishvaian MJ, Brody JR. Upgrading gemcitabine with recycled kinase inhibitors. Cell Cycle 2015; 13:2810-1. [PMID: 25486467 DOI: 10.4161/15384101.2014.954211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Fernando F Blanco
- a Department of Surgery ; Thomas Jefferson University ; Philadelphia , PA USA
| | | | | |
Collapse
|
68
|
Wen Y, Li XY, Luo J. Regulatory effect of Wee1 on proliferation of colorectal cancer cells. Shijie Huaren Xiaohua Zazhi 2015; 23:5164-5170. [DOI: 10.11569/wcjd.v23.i32.5164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of Wee1 and phosphorylated Wee1 protein in colorectal cancer and normal tissues, and analyze the regulatory effect of Wee1 on cell proliferation in the human colorectal cancer cell line HCT116.
METHODS: The expression of Wee1, p-Wee1 (Ser53) and p-Wee1 (Ser642) was detected by immunohistochemistry in colorectal cancer and normal tissues. The changes in Wee1, p-Wee1 (Ser53) and p-Wee1 (Ser642) expression were detected by Western blot in starved HCT116 cells. HCT116 cells were then cultured in medium containing different concentrations of Wee1 inhibitor PD407824. Cell proliferation was determined by Cell Counting Kit-8 (CCK-8) assay, and the morphology of the cells was observed by light microscopy.
RESULTS: The positive rates of Wee1, p-Wee1 (ser53), and p-Wee1 (ser642) expression (68.00%, 85.00% and 91.00%) in colorectal cancer were significantly higher than those in normal tissues (9.62%, 21.15% and 42.31%) (P < 0.05 or P < 0.01). The expression of the Wee1 and its phosphorylated forms rose mainly at 6, 12 and 24 h. The proliferation of HCT116 cells was inhibited by Wee1 inhibitor PD407824.
CONCLUSION: The levels of Wee1 and its phosphorylation forms closely relate to the proliferation of colorectal cancer cells. Wee1 inhibitor may be a potential new treatment for colorectal cancer in the future.
Collapse
|
69
|
Affiliation(s)
- Sabine Mueller
- University of California at San Francisco, San Francisco, CA
| | | |
Collapse
|
70
|
Manic G, Obrist F, Sistigu A, Vitale I. Trial Watch: Targeting ATM-CHK2 and ATR-CHK1 pathways for anticancer therapy. Mol Cell Oncol 2015; 2:e1012976. [PMID: 27308506 PMCID: PMC4905354 DOI: 10.1080/23723556.2015.1012976] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/25/2015] [Accepted: 01/26/2015] [Indexed: 02/08/2023]
Abstract
The ataxia telangiectasia mutated serine/threonine kinase (ATM)/checkpoint kinase 2 (CHEK2, best known as CHK2) and the ATM and Rad3-related serine/threonine kinase (ATR)/CHEK1 (best known as CHK1) cascades are the 2 major signaling pathways driving the DNA damage response (DDR), a network of processes crucial for the preservation of genomic stability that act as a barrier against tumorigenesis and tumor progression. Mutations and/or deletions of ATM and/or CHK2 are frequently found in tumors and predispose to cancer development. In contrast, the ATR-CHK1 pathway is often upregulated in neoplasms and is believed to promote tumor growth, although some evidence indicates that ATR and CHK1 may also behave as haploinsufficient oncosuppressors, at least in a specific genetic background. Inactivation of the ATM-CHK2 and ATR-CHK1 pathways efficiently sensitizes malignant cells to radiotherapy and chemotherapy. Moreover, ATR and CHK1 inhibitors selectively kill tumor cells that present high levels of replication stress, have a deficiency in p53 (or other DDR players), or upregulate the ATR-CHK1 module. Despite promising preclinical results, the clinical activity of ATM, ATR, CHK1, and CHK2 inhibitors, alone or in combination with other therapeutics, has not yet been fully demonstrated. In this Trial Watch, we give an overview of the roles of the ATM-CHK2 and ATR-CHK1 pathways in cancer initiation and progression, and summarize the results of clinical studies aimed at assessing the safety and therapeutic profile of regimens based on inhibitors of ATR and CHK1, the only 2 classes of compounds that have so far entered clinics.
Collapse
Affiliation(s)
| | - Florine Obrist
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
- INSERM, UMRS1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | | | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
- Department of Biology, University of Rome “TorVergata”; Rome, Italy
| |
Collapse
|
71
|
Mazzini G, Carpignano F, Surdo S, Aredia F, Panini N, Torchio M, Erba E, Danova M, Scovassi AI, Barillaro G, Merlo S. 3D Silicon Microstructures: A New Tool for Evaluating Biological Aggressiveness of Tumor Cells. IEEE Trans Nanobioscience 2015; 14:797-805. [PMID: 26353377 DOI: 10.1109/tnb.2015.2476351] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In this work, silicon micromachined structures (SMS), consisting of arrays of 3- μ m-thick silicon walls separated by 50- μm-deep, 5- μ m-wide gaps, were applied to investigate the behavior of eight tumor cell lines, with different origins and biological aggressiveness, in a three-dimensional (3D) microenvironment. Several cell culture experiments were performed on 3D-SMS and cells grown on silicon were stained for fluorescence microscopy analyses. Most of the tumor cell lines recognized in the literature as highly aggressive (OVCAR-5, A375, MDA-MB-231, and RPMI-7951) exhibited a great ability to enter and colonize the narrow deep gaps of the SMS, whereas less aggressive cell lines (OVCAR-3, Capan-1, MCF7, and NCI-H2126) demonstrated less penetration capability and tended to remain on top of the SMS. Quantitative image analyses of several fluorescence microscopy fields of silicon samples were performed for automatic cell recognition and count, in order to quantify the fraction of cells inside the gaps, with respect to the total number of cells in the examined field. Our results show that higher fractions of cells in the gaps are obtained with more aggressive cell lines, thus supporting in a quantitative way the observation that the behavior of tumor cells on the 3D-SMS depends on their aggressiveness level.
Collapse
|
72
|
Mak JPY, Man WY, Ma HT, Poon RYC. Pharmacological targeting the ATR-CHK1-WEE1 axis involves balancing cell growth stimulation and apoptosis. Oncotarget 2015; 5:10546-57. [PMID: 25301733 PMCID: PMC4279392 DOI: 10.18632/oncotarget.2508] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 09/24/2014] [Indexed: 02/04/2023] Open
Abstract
The ATR–CHK1–WEE1 kinase cascade's functions in the DNA damage checkpoints are well established. Moreover, its roles in the unperturbed cell cycle are also increasingly being recognized. In this connection, a number of small-molecule inhibitors of ATR, CHK1, and WEE1 are being evaluated in clinical trials. Understanding precisely how cells respond to different concentrations of inhibitors is therefore of paramount importance and has broad clinical implications. Here we present evidence that in the absence of DNA damage, pharmacological inactivation of ATR was less effective in inducing mitotic catastrophe than inhibition of WEE1 and CHK1. Small-molecule inhibitors of CHK1 (AZD7762) or WEE1 (MK-1775) induced mitotic catastrophe, as characterized by dephosphorylation of CDK1Tyr15, phosphorylation of histone H3Ser10, and apoptosis. Unexpectedly, partial inhibition of WEE1 and CHK1 had the opposite effect of accelerating the cell cycle without inducing apoptosis, thereby increasing the overall cell proliferation. This was also corroborated by the finding that cell proliferation was enhanced by kinase-inactive versions of WEE1. We demonstrated that these potential limitations of the inhibitors could be overcome by targeting more than one components of the ATR–CHK1–WEE1 simultaneously. These observations reveal insights into the complex responses to pharmacological inactivation of the ATR–CHK1–WEE1 axis.
Collapse
Affiliation(s)
- Joyce P Y Mak
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Wing Yu Man
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Hoi Tang Ma
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Randy Y C Poon
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| |
Collapse
|
73
|
Targeting the Checkpoint to Kill Cancer Cells. Biomolecules 2015; 5:1912-37. [PMID: 26295265 PMCID: PMC4598780 DOI: 10.3390/biom5031912] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 08/07/2015] [Accepted: 08/11/2015] [Indexed: 12/15/2022] Open
Abstract
Cancer treatments such as radiotherapy and most of the chemotherapies act by damaging DNA of cancer cells. Upon DNA damage, cells stop proliferation at cell cycle checkpoints, which provides them time for DNA repair. Inhibiting the checkpoint allows entry to mitosis despite the presence of DNA damage and can lead to cell death. Importantly, as cancer cells exhibit increased levels of endogenous DNA damage due to an excessive replication stress, inhibiting the checkpoint kinases alone could act as a directed anti-cancer therapy. Here, we review the current status of inhibitors targeted towards the checkpoint effectors and discuss mechanisms of their actions in killing of cancer cells.
Collapse
|
74
|
Tanaka N, Patel AA, Wang J, Frederick MJ, Kalu NN, Zhao M, Fitzgerald AL, Xie TX, Silver NL, Caulin C, Zhou G, Skinner HD, Johnson FM, Myers JN, Osman AA. Wee-1 Kinase Inhibition Sensitizes High-Risk HPV+ HNSCC to Apoptosis Accompanied by Downregulation of MCl-1 and XIAP Antiapoptotic Proteins. Clin Cancer Res 2015; 21:4831-44. [PMID: 26124202 DOI: 10.1158/1078-0432.ccr-15-0279] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 06/12/2015] [Indexed: 12/21/2022]
Abstract
PURPOSE Although the majority of patients with HPV(+) oropharyngeal cancers have a favorable prognosis, there are some patients with tumors that are resistant to aggressive chemoradiotherapy with unusual patterns of locoregional and systemic recurrences. Therefore, more effective therapies are needed. In this study, we investigated the chemosensitizing efficacy of the selective Wee-1 kinase inhibitor, AZD-1775, in HPV(+) head and neck squamous cell carcinoma (HNSCC). EXPERIMENTAL DESIGN Clonogenic survival assays and an orthotopic mouse model of HPV(+) oral cancer were used to examine the in vitro and in vivo sensitivity of HPV(+) HNSCC cell lines to AZD-1775 in combination with cisplatin, respectively. Cell-cycle analysis, DNA damage (γH2AX), homologous recombination (HR), and apoptosis were examined to dissect molecular mechanisms. RESULTS We found that AZD-1775 displays single-agent activity and enhances the response of HPV(+) HNSCC cells to cisplatin both in vitro and in vivo. The sensitivity of the HPV(+) HNSCC cells to AZD-1775 alone or in combination with cisplatin was associated with G2 checkpoint abrogation, persistent DNA damage, and apoptosis induction. This finding of AZD-1775 increasing the sensitivity of HPV(+) HNSCC cells to cisplatin through apoptosis was not seen previously in the HPV(-) HNSCC cancer cells and is accompanied by a decreased expression of the antiapoptotic proteins, MCl-1and XIAP, which appear to be cleaved following AZD-1775 treatment. CONCLUSIONS AZD-1775 selectively sensitizes HPV(+) HNSCC cells and orthotopic oral xenografts to cisplatin through apoptosis and support the clinical investigation of AZD-1775 in combination with cisplatin particularly in patients with advanced and recurrent metastatic HPV(+) HNSCC tumors.
Collapse
Affiliation(s)
- Noriaki Tanaka
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ameeta A Patel
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jiping Wang
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mitchell J Frederick
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nene N Kalu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mei Zhao
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alison L Fitzgerald
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tong-xin Xie
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Natalie L Silver
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Carlos Caulin
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ge Zhou
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Heath D Skinner
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Faye M Johnson
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Abdullah A Osman
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
75
|
Magnussen GI, Emilsen E, Giller Fleten K, Engesæter B, Nähse-Kumpf V, Fjær R, Slipicevic A, Flørenes VA. Combined inhibition of the cell cycle related proteins Wee1 and Chk1/2 induces synergistic anti-cancer effect in melanoma. BMC Cancer 2015; 15:462. [PMID: 26054341 PMCID: PMC4460948 DOI: 10.1186/s12885-015-1474-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 05/26/2015] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Malignant melanoma has an increasing incidence rate and the metastatic disease is notoriously resistant to standard chemotherapy. Loss of cell cycle checkpoints is frequently found in many cancer types and makes the cells reliant on compensatory mechanisms to control progression. This feature may be exploited in therapy, and kinases involved in checkpoint regulation, such as Wee1 and Chk1/2, have thus become attractive therapeutic targets. METHODS In the present study we combined a Wee1 inhibitor (MK1775) with Chk1/2 inhibitor (AZD7762) in malignant melanoma cell lines grown in vitro (2D and 3D cultures) and in xenografts models. RESULTS Our in vitro studies showed that combined inhibition of Wee1 and Chk1/2 synergistically decreased viability and increased apoptosis (cleavage of caspase 3 and PARP), which may be explained by accumulation of DNA-damage (increased expression of γ-H2A.X)--and premature mitosis of S-phase cells. Compared to either inhibitor used as single agents, combined treatment reduced spheroid growth and led to greater tumour growth inhibition in melanoma xenografts. CONCLUSIONS These data provide a rationale for further evaluation of the combination of Wee1 and Chk1/2 inhibitors in malignant melanoma.
Collapse
Affiliation(s)
- Gry Irene Magnussen
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, N-0310, Oslo, Norway.
| | - Elisabeth Emilsen
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, N-0310, Oslo, Norway.
| | - Karianne Giller Fleten
- Department of Tumour Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo, Norway.
| | - Birgit Engesæter
- Department of Tumour Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo, Norway.
| | - Viola Nähse-Kumpf
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo, Norway.
| | - Roar Fjær
- Department of Medical Genetics, Ullevål University Hospital, Oslo, Norway.
| | - Ana Slipicevic
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, N-0310, Oslo, Norway.
| | - Vivi Ann Flørenes
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, N-0310, Oslo, Norway.
| |
Collapse
|
76
|
Dobbelstein M, Sørensen CS. Exploiting replicative stress to treat cancer. Nat Rev Drug Discov 2015; 14:405-23. [PMID: 25953507 DOI: 10.1038/nrd4553] [Citation(s) in RCA: 218] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
DNA replication in cancer cells is accompanied by stalling and collapse of the replication fork and signalling in response to DNA damage and/or premature mitosis; these processes are collectively known as 'replicative stress'. Progress is being made to increase our understanding of the mechanisms that govern replicative stress, thus providing ample opportunities to enhance replicative stress for therapeutic purposes. Rather than trying to halt cell cycle progression, cancer therapeutics could aim to increase replicative stress by further loosening the checkpoints that remain available to cancer cells and ultimately inducing the catastrophic failure of proliferative machineries. In this Review, we outline current and future approaches to achieve this, emphasizing the combination of conventional chemotherapy with targeted approaches.
Collapse
Affiliation(s)
- Matthias Dobbelstein
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences, Ernst Caspari Haus, University of Göttingen, 37077 Göttingen, Germany
| | | |
Collapse
|
77
|
Bose P, Grant S. Rational Combinations of Targeted Agents in AML. J Clin Med 2015; 4:634-664. [PMID: 26113989 PMCID: PMC4470160 DOI: 10.3390/jcm4040634] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 01/06/2015] [Indexed: 12/20/2022] Open
Abstract
Despite modest improvements in survival over the last several decades, the treatment of AML continues to present a formidable challenge. Most patients are elderly, and these individuals, as well as those with secondary, therapy-related, or relapsed/refractory AML, are particularly difficult to treat, owing to both aggressive disease biology and the high toxicity of current chemotherapeutic regimens. It has become increasingly apparent in recent years that coordinated interruption of cooperative survival signaling pathways in malignant cells is necessary for optimal therapeutic results. The modest efficacy of monotherapy with both cytotoxic and targeted agents in AML testifies to this. As the complex biology of AML continues to be elucidated, many “synthetic lethal” strategies involving rational combinations of targeted agents have been developed. Unfortunately, relatively few of these have been tested clinically, although there is growing interest in this area. In this article, the preclinical and, where available, clinical data on some of the most promising rational combinations of targeted agents in AML are summarized. While new molecules should continue to be combined with conventional genotoxic drugs of proven efficacy, there is perhaps a need to rethink traditional philosophies of clinical trial development and regulatory approval with a focus on mechanism-based, synergistic strategies.
Collapse
Affiliation(s)
- Prithviraj Bose
- Department of Internal Medicine, Virginia Commonwealth University and VCU Massey Cancer Center Center, 1201 E Marshall St, MMEC 11-213, P.O. Box 980070, Richmond, VA 23298, USA; E-Mail:
| | - Steven Grant
- Departments of Internal Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, Human and Molecular Genetics and the Institute for Molecular Medicine, Virginia Commonwealth University and VCU Massey Cancer Center, 401 College St, P.O. Box 980035, Richmond, VA 23298, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-804-828-5211; Fax: +1-804-628-5920
| |
Collapse
|
78
|
Aleem E, Arceci RJ. Targeting cell cycle regulators in hematologic malignancies. Front Cell Dev Biol 2015; 3:16. [PMID: 25914884 PMCID: PMC4390903 DOI: 10.3389/fcell.2015.00016] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/25/2015] [Indexed: 12/20/2022] Open
Abstract
Hematologic malignancies represent the fourth most frequently diagnosed cancer in economically developed countries. In hematologic malignancies normal hematopoiesis is interrupted by uncontrolled growth of a genetically altered stem or progenitor cell (HSPC) that maintains its ability of self-renewal. Cyclin-dependent kinases (CDKs) not only regulate the mammalian cell cycle, but also influence other vital cellular processes, such as stem cell renewal, differentiation, transcription, epigenetic regulation, apoptosis, and DNA repair. Chromosomal translocations, amplification, overexpression and altered CDK activities have been described in different types of human cancer, which have made them attractive targets for pharmacological inhibition. Mouse models deficient for one or more CDKs have significantly contributed to our current understanding of the physiological functions of CDKs, as well as their roles in human cancer. The present review focuses on selected cell cycle kinases with recent emerging key functions in hematopoiesis and in hematopoietic malignancies, such as CDK6 and its role in MLL-rearranged leukemia and acute lymphocytic leukemia, CDK1 and its regulator WEE-1 in acute myeloid leukemia (AML), and cyclin C/CDK8/CDK19 complexes in T-cell acute lymphocytic leukemia. The knowledge gained from gene knockout experiments in mice of these kinases is also summarized. An overview of compounds targeting these kinases, which are currently in clinical development in various solid tumors and hematopoietic malignances, is presented. These include the CDK4/CDK6 inhibitors (palbociclib, LEE011, LY2835219), pan-CDK inhibitors that target CDK1 (dinaciclib, flavopiridol, AT7519, TG02, P276-00, terampeprocol and RGB 286638) as well as the WEE-1 kinase inhibitor, MK-1775. The advantage of combination therapy of cell cycle inhibitors with conventional chemotherapeutic agents used in the treatment of AML, such as cytarabine, is discussed.
Collapse
Affiliation(s)
- Eiman Aleem
- Department of Child Health, The Ronald A. Matricaria Institute of Molecular Medicine at Phoenix Children's Hospital, University of Arizona College of Medicine-Phoenix Phoenix, AZ, USA ; Department of Zoology, Faculty of Science, Alexandria University Alexandria, Egypt
| | - Robert J Arceci
- Department of Child Health, The Ronald A. Matricaria Institute of Molecular Medicine at Phoenix Children's Hospital, University of Arizona College of Medicine-Phoenix Phoenix, AZ, USA
| |
Collapse
|
79
|
Aarts M, Bajrami I, Herrera-Abreu MT, Elliott R, Brough R, Ashworth A, Lord CJ, Turner NC. Functional Genetic Screen Identifies Increased Sensitivity to WEE1 Inhibition in Cells with Defects in Fanconi Anemia and HR Pathways. Mol Cancer Ther 2015; 14:865-76. [PMID: 25673822 PMCID: PMC6485454 DOI: 10.1158/1535-7163.mct-14-0845] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/04/2015] [Indexed: 01/11/2023]
Abstract
WEE1 kinase regulates CDK1 and CDK2 activity to facilitate DNA replication during S-phase and to prevent unscheduled entry into mitosis. WEE1 inhibitors synergize with DNA-damaging agents that arrest cells in S-phase by triggering direct mitotic entry without completing DNA synthesis, resulting in catastrophic chromosome fragmentation and apoptosis. Here, we investigated how WEE1 inhibition could be best exploited for cancer therapy by performing a functional genetic screen to identify novel determinants of sensitivity to WEE1 inhibition. Inhibition of kinases that regulate CDK activity, CHK1 and MYT1, synergized with WEE1 inhibition through both increased replication stress and forced mitotic entry of S-phase cells. Loss of multiple components of the Fanconi anemia (FA) and homologous recombination (HR) pathways, in particular DNA helicases, sensitized to WEE1 inhibition. Silencing of FA/HR genes resulted in excessive replication stress and nucleotide depletion following WEE1 inhibition, which ultimately led to increased unscheduled mitotic entry. Our results suggest that cancers with defects in FA and HR pathways may be targeted by WEE1 inhibition, providing a basis for a novel synthetic lethal strategy for cancers harboring FA/HR defects.
Collapse
Affiliation(s)
- Marieke Aarts
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Ilirjana Bajrami
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Maria T Herrera-Abreu
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Richard Elliott
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Rachel Brough
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Alan Ashworth
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Christopher J Lord
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Nicholas C Turner
- CRUK Gene Function Laboratory, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom. Breast Unit, Royal Marsden Hospital, London, United Kingdom.
| |
Collapse
|
80
|
Syljuåsen RG, Hasvold G, Hauge S, Helland Å. Targeting lung cancer through inhibition of checkpoint kinases. Front Genet 2015; 6:70. [PMID: 25774168 PMCID: PMC4343027 DOI: 10.3389/fgene.2015.00070] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 02/10/2015] [Indexed: 12/28/2022] Open
Abstract
Inhibitors of checkpoint kinases ATR, Chk1, and Wee1 are currently being tested in preclinical and clinical trials. Here, we review the basic principles behind the use of such inhibitors as anticancer agents, and particularly discuss their potential for treatment of lung cancer. As lung cancer is one of the most deadly cancers, new treatment strategies are highly needed. We discuss how checkpoint kinase inhibition in principle can lead to selective killing of lung cancer cells while sparing the surrounding normal tissues. Several features of lung cancer may potentially be exploited for targeting through inhibition of checkpoint kinases, including mutated p53, low ERCC1 levels, amplified Myc, tumor hypoxia and presence of lung cancer stem cells. Synergistic effects have also been reported between inhibitors of ATR/Chk1/Wee1 and conventional lung cancer treatments, such as gemcitabine, cisplatin, or radiation. Altogether, inhibitors of ATR, Chk1, and Wee1 are emerging as new cancer treatment agents, likely to be useful in lung cancer treatment. However, as lung tumors are very diverse, the inhibitors are unlikely to be effective in all patients, and more work is needed to determine how such inhibitors can be utilized in the most optimal ways.
Collapse
Affiliation(s)
- Randi G Syljuåsen
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital , Oslo, Norway
| | - Grete Hasvold
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital , Oslo, Norway
| | - Sissel Hauge
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital , Oslo, Norway
| | - Åslaug Helland
- Department of Genetics, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital , Oslo, Norway ; Department of Oncology, Norwegian Radium Hospital, Oslo University Hospital , Oslo, Norway
| |
Collapse
|
81
|
Ghiasi N, Habibagahi M, Rosli R, Ghaderi A, Yusoff K, Hosseini A, Abdullah S, Jaberipour M. Tumour suppressive effects of WEE1 gene silencing in breast cancer cells. Asian Pac J Cancer Prev 2015; 14:6605-11. [PMID: 24377575 DOI: 10.7314/apjcp.2013.14.11.6605] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND WEE1 is a G2/M checkpoint regulator protein. Various studies have indicated that WEE1 could be a good target for cancer therapy. The main aim of this study was to asssess the tumor suppressive potential of WEE1 silencing in two different breast cancer cell lines, MCF7 which carries the wild-type p53 and MDA-MB468 which contains a mutant type. MATERIALS AND METHODS After WEE1 knockdown with specific shRNAs downstream effects on cell viability and cell cycle progression were determined using MTT and flow cytometry analyses, respectively. Real-time PCR and Western blotting were conducted to assess the effect of WEE1 inhibition on the expression of apoptotic (p53) and anti-apoptotic (Bcl2) factors and also a growth marker (VEGF). RESULTS The results showed that WEE1 inhibition could cause a significant decrease in the viability of both MCF7 and MDA-MB-468 breast cancer cell lines by more than 50%. Interestingly, DNA content assays showed a significant increase in apoptotic cells following WEE1 silencing. WEE1 inhibition also induced up- regulation of the apoptotic marker, p53, in breast cancer cells. A significant decrease in the expression of VEGF and Bcl-2 was observed following WEE1 inhibition in both cell lines. CONCLUSIONS In concordance with previous studies, our data showed that WEE1 inhibition could induce G2 arrest abrogation and consequent cell death in breast cancer cells. Moreover, in this study, the observed interactions between the pro- and anti-apoptotic proteins and decrease in the angiogenesis marker expression confirm the susceptibility to apoptosis and validate the tumor suppressive effect of WEE1 inhibition in breast cancer cells. Interestingly, the levels of the sensitivity to WEE1 silencing in breast cancer cells, MCF7 and MDA-MB468, seem to be in concordance with the level of p53 expression.
Collapse
Affiliation(s)
- Naghmeh Ghiasi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran E-mail :
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Hanmod SS, Wang G, Edwards H, Buck SA, Ge Y, Taub JW, Wang Z. Targeting histone deacetylases (HDACs) and Wee1 for treating high-risk neuroblastoma. Pediatr Blood Cancer 2015; 62:52-9. [PMID: 25308916 DOI: 10.1002/pbc.25232] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 07/30/2014] [Indexed: 12/19/2022]
Abstract
BACKGROUND Despite advances in treatment regimens, patients with high-risk neuroblastoma have long-term survival rates of < 40%. Wee1 inhibition in combination with CHK1 inhibition has shown promising results in neuroblastoma cells. In addition, it has been demonstrated that panobinostat can downregulate CHK1. Therefore, combination of panobinostat and MK-1775 may result in synergistic cytotoxicity against neuroblastoma cell lines. PROCEDURE In vitro cytotoxicities of panobinostat and MK-1775 at clinically achievable concentrations, either alone or in combination, were evaluated in SK-N-AS, SK-N-DZ, and SK-N-BE(2) high-risk neuroblastoma cell lines using MTT assays. The mechanism of antitumor interaction was investigated using propidium iodide (PI) staining and flow cytometry analysis to determine apoptosis, as well as Western blotting to assess expression of phosphorylated CDK1/2, CHK1, and H2AX. RESULTS Treatment of neuroblastoma cell lines with 500 nM MK-1775 caused growth arrest and apoptosis in SK-N-DZ and SK-N-AS, while it had minimal effect on the SK-N-BE(2) cell line. The combination of panobinostat and MK-1775 resulted in synergistic antitumor interactions in all three of the cell lines tested. MK-1775 treatment in SK-N-BE(2) cells induced increased levels of p-CHK1(S345) , which could be decreased by the addition of panobinostat. This was accompanied by increased DNA damage and apoptosis. CONCLUSIONS The combination of panobinostat and MK-1775 has synergistic antitumor activity against neuroblastoma cell lines and holds promise as a potential treatment strategy for the management of high-risk neuroblastoma patients.
Collapse
Affiliation(s)
- Santosh S Hanmod
- Division of Pediatric Hematology/Oncology, Children's Hospital of Michigan, Detroit, Michigan
| | | | | | | | | | | | | |
Collapse
|
83
|
Wang G, Niu X, Zhang W, Caldwell JT, Edwards H, Chen W, Taub JW, Zhao L, Ge Y. Synergistic antitumor interactions between MK-1775 and panobinostat in preclinical models of pancreatic cancer. Cancer Lett 2014; 356:656-68. [PMID: 25458954 DOI: 10.1016/j.canlet.2014.10.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 09/26/2014] [Accepted: 10/14/2014] [Indexed: 01/05/2023]
Abstract
Pancreatic cancer remains a clinical challenge, thus new therapies are urgently needed. The selective Wee1 inhibitor MK-1775 has demonstrated promising results when combined with DNA damaging agents, and more recently with CHK1 inhibitors in various malignancies. We have previously demonstrated that treatment with the pan-histone deacetylase inhibitor panobinostat (LBH589) can cause down-regulation of CHK1. Accordingly, we investigated using panobinostat to down-regulate CHK1 in combination with MK-1775 to enhance cell death in preclinical pancreatic cancer models. We demonstrate that MK-1775 treatment results in increased H2AX phosphorylation, indicating increased DNA double-strand breaks, and activation of CHK1, which are both dependent on CDK activity. Combination of MK-1775 and panobinostat resulted in synergistic antitumor activity in six pancreatic cancer cell lines. Finally, our in vivo study using a pancreatic xenograft model reveals promising cooperative antitumor activity between MK-1775 and panobinostat. Our study provides compelling evidence that the combination of MK-1775 and panobinostat has antitumor activity in preclinical models of pancreatic cancer and supports the clinical development of panobinostat in combination with MK-1775 for the treatment of this deadly disease.
Collapse
Affiliation(s)
- Guan Wang
- 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; Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaojia Niu
- 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
- 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
| | - J Timothy Caldwell
- MD/PhD Program, Wayne State University School of Medicine, Detroit, MI, USA; Cancer Biology Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - 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
| | - Wei Chen
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jeffrey W Taub
- Department of Pediatrics, 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
| | - Lijing Zhao
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China.
| | - Yubin Ge
- 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; 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
|
84
|
A regimen combining the Wee1 inhibitor AZD1775 with HDAC inhibitors targets human acute myeloid leukemia cells harboring various genetic mutations. Leukemia 2014; 29:807-18. [PMID: 25283841 PMCID: PMC4387110 DOI: 10.1038/leu.2014.296] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/09/2014] [Accepted: 09/22/2014] [Indexed: 02/05/2023]
Abstract
AZD1775 targets the cell cycle checkpoint kinase Wee1 and potentiates genotoxic agent cytotoxicity through p53-dependent or -independent mechanisms. Here, we report that AZD1775 interacted synergistically with histone deacetylase inhibitors (HDACIs e.g., Vorinostat), which interrupt the DNA damage response (DDR), to kill p53-wild type or -deficient as well as FLT3-ITD leukemia cells in association with pronounced Wee1 inhibition and diminished cdc2/Cdk1 Y15 phosphorylation. Similarly, Wee1 shRNA knock-down significantly sensitized cells to HDACIs. While AZD1775 induced Chk1 activation, reflected by markedly increased Chk1 S296/S317/S345 phosphorylation leading to inhibitory T14 phosphorylation of cdc2/Cdk1, these compensatory responses were sharply abrogated by HDACIs. This was accompanied by premature mitotic entry, multiple mitotic abnormalities, and accumulation of early S-phase cells displaying increased newly replicated DNA, culminating in robust DNA damage and apoptosis. The regimen was active against patient-derived AML cells harboring either wild type or mutant p53, and various NGS-defined mutations. Primitive CD34+/CD123+/CD38− populations enriched for leukemia-initiating progenitors, but not normal CD34+ hematopoietic cells, were highly susceptible to this regimen. Finally, combining AZD1775 with Vorinostat in AML murine xenografts significantly reduced tumor burden and prolonged animal survival. A strategy combining Wee1 with HDACI inhibition warrants further investigation in AML with poor prognostic genetic aberrations.
Collapse
|
85
|
Karnak D, Engelke CG, Parsels LA, Kausar T, Wei D, Robertson JR, Marsh KB, Davis MA, Zhao L, Maybaum J, Lawrence TS, Morgan MA. Combined inhibition of Wee1 and PARP1/2 for radiosensitization in pancreatic cancer. Clin Cancer Res 2014; 20:5085-96. [PMID: 25117293 DOI: 10.1158/1078-0432.ccr-14-1038] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE While the addition of radiation to chemotherapy improves survival in patients with locally advanced pancreatic cancer, more effective therapies are urgently needed. Thus, we investigated the radiosensitizing efficacy of the novel drug combination of Wee1 and PARP1/2 inhibitors (AZD1775 and olaparib, respectively) in pancreatic cancer. EXPERIMENTAL DESIGN Radiosensitization of AsPC-1 or MiaPaCa-2 human pancreatic cancer cells was assessed by clonogenic survival and tumor growth assays. Mechanistically, the effects of AZD1775, olaparib, and radiation on cell cycle, DNA damage (γH2AX), and homologous recombination repair (HRR) were determined. RESULTS Treatment of AsPC-1 and MiaPaCa-2 cells with either AZD1775 or olaparib caused modest radiosensitization, whereas treatment with the combination significantly increased radiosensitization. Radiosensitization by the combination of AZD1775 and olaparib was associated with G2 checkpoint abrogation and persistent DNA damage. In addition, AZD1775 inhibited HRR activity and prevented radiation-induced Rad51 focus formation. Finally, in vivo, in MiaPaCa-2-derived xenografts, olaparib did not radiosensitize, whereas AZD1775 produced moderate, yet significant, radiosensitization (P < 0.05). Importantly, the combination of AZD1775 and olaparib produced highly significant radiosensitization (P < 0.0001) evidenced by a 13-day delay in tumor volume doubling (vs. radiation alone) and complete eradication of 20% of tumors. CONCLUSIONS Taken together, these results demonstrate the efficacy of combined inhibition of Wee1 and PARP inhibitors for radiosensitizing pancreatic cancers and support the model that Wee1 inhibition sensitizes cells to PARP inhibitor-mediated radiosensitization through inhibition of HRR and abrogation of the G2 checkpoint, ultimately resulting in unrepaired, lethal DNA damage and radiosensitization. Clin Cancer Res; 20(19); 5085-96. ©2014 AACR.
Collapse
Affiliation(s)
- David Karnak
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Carl G Engelke
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Leslie A Parsels
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Tasneem Kausar
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Dongping Wei
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jordan R Robertson
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Katherine B Marsh
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Mary A Davis
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Lili Zhao
- Biostatistics Unit, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Jonathan Maybaum
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Meredith A Morgan
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan.
| |
Collapse
|
86
|
Qi W, Xie C, Li C, Caldwell JT, Edwards H, Taub JW, Wang Y, Lin H, Ge Y. CHK1 plays a critical role in the anti-leukemic activity of the wee1 inhibitor MK-1775 in acute myeloid leukemia cells. J Hematol Oncol 2014; 7:53. [PMID: 25084614 PMCID: PMC4237862 DOI: 10.1186/s13045-014-0053-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 07/08/2014] [Indexed: 11/24/2022] Open
Abstract
Background Acute myeloid leukemia (AML) remains a difficult disease to treat and requires new therapies to improve treatment outcome. Wee1 inhibitors have been used to prevent activation of the G2 cell cycle checkpoint, thus enhancing the antitumor activity of DNA damaging agents. In this study, we investigated MK-1775 in AML cell lines and diagnostic blast samples to identify sensitive subtypes as well as possible mechanisms of resistance. Methods In vitro MK-1775 cytotoxicities of AML cell lines and diagnostic blasts were measured using MTT assays. The effects of MK-1775 on cell cycle progression and related proteins were determined by propidium iodide (PI) staining and flow cytometry analysis and Western blotting. Drug-induced apoptosis was determined using annexin V/PI staining and flow cytometry analysis. Results We found that newly diagnosed and relapsed patient samples were equally sensitive to MK-1775. In addition, patient samples harboring t(15;17) translocation were significantly more sensitive to MK-1775 than non-t(15;17) samples. MK-1775 induced apoptosis in both AML cell lines and diagnostic blast samples, accompanied by decreased phosphorylation of CDK1 and CDK2 on Tyr-15 and increased DNA double-strand breaks (DSBs). Time-course experiments, using AML cell lines, revealed a time-dependent increase in DNA DSBs, activation of CHK1 and subsequent apoptosis following MK-1775 treatment, which could be attenuated by a CDK1/2 inhibitor, Roscovitine. Simultaneous inhibition of CHK1 and Wee1 resulted in synergistic anti-leukemic activity in both AML cell lines and primary patient samples ex vivo. Conclusions Our study provides compelling evidence that CHK1 plays a critical role in the anti-leukemic activity of MK-1775 and highlights a possible mechanism of resistance to MK-1775. In addition, our study strongly supports the use of MK-1775 to treat both newly diagnosed and relapsed AML, especially cases with t(15;17) translocation, and supports the development of combination therapies with CHK1 inhibitors.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yue Wang
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology & Engineering, the Ministry of Education, and School of Life Sciences, Jilin University, Changchun, China.
| | | | | |
Collapse
|
87
|
Lal S, Burkhart RA, Beeharry N, Bhattacharjee V, Londin ER, Cozzitorto JA, Romeo C, Jimbo M, Norris ZA, Yeo CJ, Sawicki JA, Winter JM, Rigoutsos I, Yen TJ, Brody JR. HuR posttranscriptionally regulates WEE1: implications for the DNA damage response in pancreatic cancer cells. Cancer Res 2014; 74:1128-40. [PMID: 24536047 DOI: 10.1158/0008-5472.can-13-1915] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
HuR (ELAV1), an RNA-binding protein abundant in cancer cells, primarily resides in the nucleus, but under specific stress (e.g., gemcitabine), HuR translocates to the cytoplasm in which it tightly modulates the expression of mRNA survival cargo. Here, we demonstrate for the first time that stressing pancreatic ductal adenocarcinoma (PDA) cells by treatment with DNA-damaging anticancer agents (mitomycin C, oxaliplatin, cisplatin, carboplatin, and a PARP inhibitor) results in HuR's translocation from the nucleus to the cytoplasm. Importantly, silencing HuR in PDA cells sensitized the cells to these agents, whereas overexpressing HuR caused resistance. HuR's role in the efficacy of DNA-damaging agents in PDA cells was, in part, attributed to the acute upregulation of WEE1 by HuR. WEE1, a mitotic inhibitor kinase, regulates the DNA damage repair pathway, and therapeutic inhibition of WEE1 in combination with chemotherapy is currently in early phase trials for the treatment of cancer. We validate WEE1 as a HuR target in vitro and in vivo by demonstrating (i) direct binding of HuR to WEE1's mRNA (a discrete 56-bp region residing in the 3' untranslated region) and (ii) HuR siRNA silencing and overexpression directly affects the protein levels of WEE1, especially after DNA damage. HuR's positive regulation of WEE1 increases γ-H2AX levels, induces Cdk1 phosphorylation, and promotes cell-cycle arrest at the G2-M transition. We describe a novel mechanism that PDA cells use to protect against DNA damage in which HuR posttranscriptionally regulates the expression and downstream function of WEE1 upon exposure to DNA-damaging agents.
Collapse
Affiliation(s)
- Shruti Lal
- Authors' Affiliations: Department of Surgery, Division of Surgical Research, Jefferson Pancreas, Biliary and Related Cancer Center, Jefferson Medical College; Computational Medicine Center; Kimmel Cancer Center, Thomas Jefferson University; Fox Chase Cancer Center, Philadelphia; and Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Pitts TM, Davis SL, Eckhardt SG, Bradshaw-Pierce EL. Targeting nuclear kinases in cancer: development of cell cycle kinase inhibitors. Pharmacol Ther 2013; 142:258-69. [PMID: 24362082 DOI: 10.1016/j.pharmthera.2013.12.010] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 11/27/2013] [Indexed: 12/13/2022]
Abstract
Cellular proliferation is a tightly controlled set of events that is regulated by numerous nuclear protein kinases. The proteins involved include checkpoint kinases (CHK), cyclin-dependent kinases (CDK), which regulate the cell cycle and aurora kinases (AURK) and polo-like kinases (PLK), which regulate mitosis. In cancer, these nuclear kinases are often dysregulated and cause uncontrolled cell proliferation and growth. Much work has gone into developing novel therapeutics that target each of these protein kinases in cancer but none have been approved in patients. In this review we provide an overview of the current compounds being developed clinically to target these nuclear kinases involved in regulating the cell cycle and mitosis.
Collapse
Affiliation(s)
- Todd M Pitts
- Division of Medical Oncology, University of Colorado Denver, Anschutz Medical Campus, United States; University of Colorado Cancer Center, University of Colorado Denver, Anschutz Medical Campus, United States.
| | - S Lindsey Davis
- Division of Medical Oncology, University of Colorado Denver, Anschutz Medical Campus, United States
| | - S Gail Eckhardt
- Division of Medical Oncology, University of Colorado Denver, Anschutz Medical Campus, United States; University of Colorado Cancer Center, University of Colorado Denver, Anschutz Medical Campus, United States
| | - Erica L Bradshaw-Pierce
- Department of Pharmaceutical Sciences, University of Colorado Denver, Anschutz Medical Campus, United States; University of Colorado Cancer Center, University of Colorado Denver, Anschutz Medical Campus, United States
| |
Collapse
|
89
|
Rohe A, Henze C, Erdmann F, Sippl W, Schmidt M. A fluorescence anisotropy-based Myt1 kinase binding assay. Assay Drug Dev Technol 2013; 12:136-44. [PMID: 24229357 DOI: 10.1089/adt.2013.534] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Abstract The human Myt1 kinase is a regulator of Cdk1/CycB and, hence, important for the G2/M transition in the cell cycle. It may act as a target for drug development, but suitable assay systems for assessing potential inhibitors are lacking so far. Herein, we describe the rational development of a fluorescence anisotropy-based kinase binding assay. A suitable fluoroprobe based on the tyrosine kinase inhibitor dasatinib was synthesized and tested with Myt1 and several other kinases for control purposes. The probe acted as expected in terms of specificity and reversibility, and a Myt1 assay was set up. Notwithstanding the moderate Kd of the starting compound dasatinib before chemical modification, satisfying Z' factors >0.5 were achieved. A validation with known kinase inhibitors demonstrated the applicability of the assay and led to a reliable ranking of the tested active compounds.
Collapse
Affiliation(s)
- Alexander Rohe
- 1 Department of Medicinal Chemistry, Martin Luther University Halle-Wittenberg , Halle, Germany
| | | | | | | | | |
Collapse
|
90
|
Chaudhuri L, Vincelette ND, Koh BD, Naylor RM, Flatten KS, Peterson KL, McNally A, Gojo I, Karp JE, Mesa RA, Sproat LO, Bogenberger JM, Kaufmann SH, Tibes R. CHK1 and WEE1 inhibition combine synergistically to enhance therapeutic efficacy in acute myeloid leukemia ex vivo. Haematologica 2013; 99:688-96. [PMID: 24179152 DOI: 10.3324/haematol.2013.093187] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Novel combinations targeting new molecular vulnerabilities are needed to improve the outcome of patients with acute myeloid leukemia. We recently identified WEE1 kinase as a novel target in leukemias. To identify genes that are synthetically lethal with WEE1 inhibition, we performed a short interfering RNA screen directed against cell cycle and DNA repair genes during concurrent treatment with the WEE1 inhibitor MK1775. CHK1 and ATR, genes encoding two replication checkpoint kinases, were among the genes whose silencing enhanced the effects of WEE1 inhibition most, whereas CDK2 short interfering RNA antagonized MK1775 effects. Building on this observation, we examined the impact of combining MK1775 with selective small molecule inhibitors of CHK1, ATR and cyclin-dependent kinases. The CHK1 inhibitor MK8776 sensitized acute myeloid leukemia cell lines and primary leukemia specimens to MK1775 ex vivo, whereas smaller effects were observed with the MK1775/MK8776 combination in normal myeloid progenitors. The ATR inhibitor VE-821 likewise enhanced the antiproliferative effects of MK1775, whereas the cyclin-dependent kinase inhibitor roscovitine antagonized MK1775. Further studies showed that MK8776 enhanced MK1775-mediated activation of the ATR/CHK1 pathway in acute leukemia cell lines and ex vivo. These results indicate that combined cell cycle checkpoint interference with MK1775/MK8776 warrants further investigation as a potential treatment for acute myeloid leukemia.
Collapse
|
91
|
McNeely S, Beckmann R, Bence Lin AK. CHEK again: revisiting the development of CHK1 inhibitors for cancer therapy. Pharmacol Ther 2013; 142:1-10. [PMID: 24140082 DOI: 10.1016/j.pharmthera.2013.10.005] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 10/05/2013] [Indexed: 02/06/2023]
Abstract
CHEK1 encodes the serine/threonine kinase CHK1, a central component of the DNA damage response. CHK1 regulates cell cycle checkpoints following genotoxic stress to prevent the entry of cells with damaged DNA into mitosis and coordinates various aspects of DNA repair. Accordingly, CHK1 has become a target of considerable interest in oncology. CHK1 inhibitors potentiate the efficacy of DNA-damaging chemotherapeutics by abrogating CHK1-mediated cell cycle arrest and preventing repair of damaged DNA. In addition, CHK1 inhibitors interfere with the biological role of CHK1 as a principal regulator of the cell cycle that controls the initiation of DNA replication, stabilizes replication forks, and coordinates mitosis. Since these functions of CHK1 facilitate progression through an unperturbed cell cycle, CHK1 inhibitors are being developed not only as chemopotentiators, but also as single-agent therapies. This review is intended to provide information on the current progress of CHK1 inhibitors in pre-clinical and clinical development and will focus on mechanisms of single-agent activity and potential strategies for patient tailoring and combinations with non-genotoxic agents.
Collapse
Affiliation(s)
- S McNeely
- Eli Lilly and Company, Indianapolis, IN, United States.
| | - R Beckmann
- Eli Lilly and Company, Indianapolis, IN, United States
| | - A K Bence Lin
- Eli Lilly and Company, Indianapolis, IN, United States
| |
Collapse
|
92
|
Chen B, Duan L, Yin G, Tan J, Jiang X. Simultaneously expressed miR-424 and miR-381 synergistically suppress the proliferation and survival of renal cancer cells---Cdc2 activity is up-regulated by targeting WEE1. Clinics (Sao Paulo) 2013; 68:825-33. [PMID: 23778472 PMCID: PMC3674285 DOI: 10.6061/clinics/2013(06)17] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Accepted: 02/26/2013] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES MiRNAs are intrinsic RNAs that interfere with protein translation. Few studies on the synergistic effects of miRNAs have been reported. Both miR-424 and miR-381 have been individually reported to be involved in carcinogenesis. They share a common putative target, WEE1, which is described as an inhibitor of G2/M progression. Here, we studied the synergistic effects of miR-424 and miR-381 on renal cancer cells. METHODS The viability of 786-O cells was analyzed after transfection with either a combination of miR-424 and miR-381 or each miRNA alone. We investigated cell cycle progression and apoptosis with flow cytometry. To confirm apoptosis and the abrogation of G2/M arrest, we determined the level of pHH3, which is an indicator of mitosis, and caspase-3/7 activity. The expression levels of WEE1, Cdc25, γH2AX, and Cdc2 were manipulated to investigate the roles of these proteins in the miRNA-induced anti-tumor effects. To verify that WEE1 was a direct target of both miR-424 and miR-381, we performed a dual luciferase reporter assay. RESULTS We showed that the combination of these miRNAs synergistically inhibited proliferation, abrogated G2/M arrest, and induced apoptosis. This combination led to Cdc2 activation through WEE1 inhibition. This regulation was more effective when cells were treated with both miRNAs than with either miRNA alone, indicating synergy between these miRNAs. WEE1 was verified to be a direct target of each miRNA according to the luciferase reporter assay. CONCLUSIONS These data clearly demonstrate that these two miRNAs might synergistically act as novel modulators of tumorigenesis by down-regulating WEE1 expression in renal cell cancer cells.
Collapse
Affiliation(s)
- Binghai Chen
- Third Xiang-Ya Hospital of Central South University, Department of Urology, Changsha, Hunan/China
| | | | | | | | | |
Collapse
|
93
|
WEE1 inhibition and genomic instability in cancer. Biochim Biophys Acta Rev Cancer 2013; 1836:227-35. [PMID: 23727417 DOI: 10.1016/j.bbcan.2013.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 05/21/2013] [Accepted: 05/23/2013] [Indexed: 01/23/2023]
Abstract
One of the hallmarks of cancer is genomic instability controlled by cell cycle checkpoints. The G1 and G2 checkpoints allow DNA damage responses, whereas the mitotic checkpoint enables correct seggregation of the sister chromosomes to prevent aneuploidy. Cancer cells often lack a functional G1 arrest and rely on G2 arrest for DNA damage responses. WEE1 kinase is an important regulator of the G2 checkpoint and is overexpressed in various cancer types. Inhibition of WEE1 is a promising strategy in cancer therapy in combination with DNA-damaging agents, especially when cancer cells harbor p53 mutations, as it causes mitotic catastrophy when DNA is not repaired during G2 arrest. Cancer cell response to WEE1 inhibition monotherapy has also been demonstrated in various types of cancer, including p53 wild-type cancers. We postulate that chromosomal instability can explain tumor response to WEE1 monotherapy. Therefore, chromosomal instability may need to be taken into account when determining the most effective strategy for the use of WEE1 inhibitors in cancer therapy.
Collapse
|
94
|
Guertin AD, Li J, Liu Y, Hurd MS, Schuller AG, Long B, Hirsch HA, Feldman I, Benita Y, Toniatti C, Zawel L, Fawell SE, Gilliland DG, Shumway SD. Preclinical evaluation of the WEE1 inhibitor MK-1775 as single-agent anticancer therapy. Mol Cancer Ther 2013; 12:1442-52. [PMID: 23699655 DOI: 10.1158/1535-7163.mct-13-0025] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Inhibition of the DNA damage checkpoint kinase WEE1 potentiates genotoxic chemotherapies by abrogating cell-cycle arrest and proper DNA repair. However, WEE1 is also essential for unperturbed cell division in the absence of extrinsic insult. Here, we investigate the anticancer potential of a WEE1 inhibitor, independent of chemotherapy, and explore a possible cellular context underlying sensitivity to WEE1 inhibition. We show that MK-1775, a potent and selective ATP-competitive inhibitor of WEE1, is cytotoxic across a broad panel of tumor cell lines and induces DNA double-strand breaks. MK-1775-induced DNA damage occurs without added chemotherapy or radiation in S-phase cells and relies on active DNA replication. At tolerated doses, MK-1775 treatment leads to xenograft tumor growth inhibition or regression. To begin addressing potential response markers for MK-1775 monotherapy, we focused on PKMYT1, a kinase functionally related to WEE1. Knockdown of PKMYT1 lowers the EC(50) of MK-1775 by five-fold but has no effect on the cell-based response to other cytotoxic drugs. In addition, knockdown of PKMYT1 increases markers of DNA damage, γH2AX and pCHK1(S345), induced by MK-1775. In a post hoc analysis of 305 cell lines treated with MK-1775, we found that expression of PKMYT1 was below average in 73% of the 33 most sensitive cell lines. Our findings provide rationale for WEE1 inhibition as a potent anticancer therapy independent of a genotoxic partner and suggest that low PKMYT1 expression could serve as an enrichment biomarker for MK-1775 sensitivity.
Collapse
Affiliation(s)
- Amy D Guertin
- Oncology Biology, Merck Research Laboratories, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Matthews TP, Jones AM, Collins I. Structure-based design, discovery and development of checkpoint kinase inhibitors as potential anticancer therapies. Expert Opin Drug Discov 2013; 8:621-40. [PMID: 23594139 DOI: 10.1517/17460441.2013.788496] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Checkpoint kinase (CHK) inhibitors offer the promise of enhancing the effectiveness of widely prescribed cancer chemotherapies and radiotherapy by inhibiting the DNA damage response, as well as the potential for single agent efficacy. AREAS COVERED This article surveys structural insights into the checkpoint kinases CHK1 and CHK2 that have been exploited to enhance the selectivity and potency of small molecule inhibitors. Furthermore, the authors review the use of mechanistic cellular assays to guide the optimisation of inhibitors. Finally, the authors discuss the status of the current clinical candidates and emerging new clinical contexts for CHK1 and CHK2 inhibitors, including the prospects for single agent efficacy. EXPERT OPINION Protein-bound water molecules play key roles in structural features that can be targeted to gain high selectivity for either enzyme. The results of early phase clinical trials of checkpoint inhibitors have been mixed, but significant progress has been made in testing the combination of CHK1 inhibitors with genotoxic chemotherapy. Second-generation CHK1 inhibitors are likely to benefit from increased selectivity and oral bioavailability. While the optimum therapeutic context for CHK2 inhibition remains unclear, the emergence of single agent preclinical efficacy for CHK1 inhibitors in specific tumour types exhibiting constitutive replication stress represents exciting progress in exploring the therapeutic potential of these agents.
Collapse
Affiliation(s)
- Thomas P Matthews
- Institute of Cancer Research, Cancer Research UK Cancer Therapeutics Unit, London SM2 5NG, UK
| | | | | |
Collapse
|
96
|
Chilà R, Celenza C, Lupi M, Damia G, Carrassa L. Chk1-Mad2 interaction: a crosslink between the DNA damage checkpoint and the mitotic spindle checkpoint. Cell Cycle 2013; 12:1083-90. [PMID: 23454898 DOI: 10.4161/cc.24090] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Chk1 is implicated in several checkpoints of the cell cycle acting as a key player in the signal transduction pathway activated in response to DNA damage and crucial for the maintenance of genomic stability. Chk1 also plays a role in the mitotic spindle checkpoint, which ensures the fidelity of mitotic segregation during mitosis, preventing chromosomal instability and aneuploidy. Mad2 is one of the main mitotic checkpoint components and also exerts a role in the cellular response to DNA damage. To investigate a possible crosslink existing between Chk1 and Mad2, we studied Mad2 protein levels after Chk1 inhibition either by specific siRNAs or by a specific and selective Chk1 inhibitor (PF-00477736), and we found that after Chk1 inhibition, Mad2 protein levels decrease only in tumor cells sensitive to Chk1 depletion. We then mapped six Chk1's phosphorylatable sites on Mad2 protein, and found that Chk1 is able to phosphorylate Mad2 in vitro on more than one site, while it is incapable of phoshorylating the Mad2 form mutated on all six phosphorylatable sites. Moreover our studies demonstrate that Chk1 co-localizes and physically associates with Mad2 in cells both under unstressed conditions and after DNA damage, thus providing new and interesting evidence on Chk1 and Mad2 crosstalk in the DNA damage checkpoint and in the mitotic spindle checkpoint.
Collapse
Affiliation(s)
- Rosaria Chilà
- Laboratory of Molecular Pharmacology, Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | | | | | | | | |
Collapse
|
97
|
Kovacikova I, Polakova S, Benko Z, Cipak L, Zhang L, Rumpf C, Miadokova E, Gregan J. A knockout screen for protein kinases required for the proper meiotic segregation of chromosomes in the fission yeast Schizosaccharomyces pombe. Cell Cycle 2013; 12:618-24. [PMID: 23370392 PMCID: PMC3594262 DOI: 10.4161/cc.23513] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The reduction of chromosome number during meiosis is achieved by two successive rounds of chromosome segregation after just single round of DNA replication. To identify novel proteins required for the proper segregation of chromosomes during meiosis, we analyzed the consequences of deleting Schizosaccharomyces pombe genes predicted to encode protein kinases that are not essential for cell viability. We show that Mph1, a member of the Mps1 family of spindle assembly checkpoint kinases, is required to prevent meiosis I homolog non-disjunction. We also provide evidence for a novel function of Spo4, the fission yeast ortholog of Dbf4-dependent Cdc7 kinase, in regulating the length of anaphase II spindles. In the absence of Spo4, abnormally elongated anaphase II spindles frequently overlap and thus destroy the linear order of nuclei in the ascus. Our observation that the spo4Δ mutant phenotype can be partially suppressed by inhibiting Cdc2-as suggests that dysregulation of the activity of this cyclin-dependent kinase may cause abnormal elongation of anaphase II spindles in spo4Δ mutant cells.
Collapse
Affiliation(s)
- Ines Kovacikova
- Max F. Perutz Laboratories, Department of Chromosome Biology, University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
98
|
Unique functions of CHK1 and WEE1 underlie synergistic anti-tumor activity upon pharmacologic inhibition. Cancer Cell Int 2012; 12:45. [PMID: 23148684 PMCID: PMC3517755 DOI: 10.1186/1475-2867-12-45] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 11/04/2012] [Indexed: 11/12/2022] Open
Abstract
Background Inhibition of kinases involved in the DNA damage response sensitizes cells to genotoxic agents by abrogating checkpoint-induced cell cycle arrest. CHK1 and WEE1 act in a pathway upstream of CDK1 to inhibit cell cycle progression in response to damaged DNA. Therapeutic targeting of either CHK1 or WEE1, in combination with chemotherapy, is under clinical evaluation. These studies examine the overlap and potential for synergy when CHK1 and WEE1 are inhibited in cancer cell models. Methods Small molecules MK-8776 and MK-1775 were used to selectively and potently inhibit CHK1 and WEE1, respectively. Results In vitro, the combination of MK-8776 and MK-1775 induces up to 50-fold more DNA damage than either MK-8776 or MK-1775 alone at a fixed concentration. This requires aberrant cyclin-dependent kinase activity but does not appear to be dependent on p53 status alone. Furthermore, DNA damage takes place primarily in S-phase cells, implying disrupted DNA replication. When dosed together, the combination of MK-8776 and MK-1775 induced more intense and more durable DNA damage as well as anti-tumor efficacy than either MK-8776 or MK-1775 dosed alone. DNA damage induced by the combination was detected in up to 40% of cells in a treated xenograft tumor model. Conclusions These results highlight the roles of WEE1 and CHK1 in maintaining genomic integrity. Importantly, the strong synergy observed upon inhibition of both kinases suggests unique yet complimentary anti-tumor effects of WEE1 and CHK1 inhibition. This demonstration of DNA double strand breaks in the absence of a DNA damaging chemotherapeutic provides preclinical rationale for combining WEE1 and CHK1 inhibitors as a cancer treatment regimen.
Collapse
|
99
|
Gillespie DA. Short-circuiting the cell cycle for cancer therapy. Cell Cycle 2012; 11:2777-8. [PMID: 22801540 PMCID: PMC3419054 DOI: 10.4161/cc.21392] [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] [Indexed: 12/02/2022] Open
Abstract
Comment on: Carrassa L, et al. Cell Cycle 2012; 2507-17.
Collapse
|