1
|
Wang Y, Xu C, Jiang Y, Tu Z, Yan J, Guo L, Dong C, Liu J, Yang X, Wang Z, Lu T, Feng J, Chen Y. Advanced Design, Synthesis, and Evaluation of Highly Selective Wee1 Inhibitors: Enhancing Pharmacokinetics and Antitumor Efficacy. J Med Chem 2024; 67:9927-9949. [PMID: 38847373 DOI: 10.1021/acs.jmedchem.3c02434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Wee1 is a kinase that regulates cell cycle arrest in response to DNA damage. Wee1 inhibition is a potential strategy to suppress the growth of tumors with defective p53 or DNA repair pathways. However, the development of Wee1 inhibitors faces some challenges. AZD1775, the first-in-class Wee1 inhibitor, has poor kinase selectivity and dose-limiting toxicity. Here, we report the discovery of 12h, a highly selective and potent Wee1 inhibitor with a favorable pharmacokinetic profile. 12h showed strong antiproliferative effects against Lovo cells, a colorectal cancer cell line, both in vitro and in vivo. Moreover, 12h showed a clean kinase profile and effectively induced cell apoptosis. Our results suggest that 12h is a promising drug candidate for further development as a novel anticancer agent.
Collapse
Affiliation(s)
- Yong Wang
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, P.R. China
| | - Chunyue Xu
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, P.R. China
| | - Yiqing Jiang
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, P.R. China
| | - Zhenlin Tu
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, P.R. China
| | - Jingxue Yan
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, P.R. China
| | - Leyi Guo
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, P.R. China
| | - Chao Dong
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, P.R. China
| | - Jiaqi Liu
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, P.R. China
| | - Xiulong Yang
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, P.R. China
| | - Ziyi Wang
- Schcool of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, P.R. China
| | - Tao Lu
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, P.R. China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, P.R. China
| | - Jie Feng
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, P.R. China
| | - Yadong Chen
- School of Sciences, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, P.R. China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, P.R. China
| |
Collapse
|
2
|
Construction and Validation of a Newly Prognostic Signature for CRISPR-Cas9-Based Cancer Dependency Map Genes in Breast Cancer. JOURNAL OF ONCOLOGY 2022; 2022:4566577. [PMID: 35096059 PMCID: PMC8791742 DOI: 10.1155/2022/4566577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022]
Abstract
Cancer Dependency Map (CDM) genes comprise an extensive series of genome-scale RNAi-based loss-of-function tests; hence, it served as a method based on the CRISPR-Cas9 technique that could assist scientists in investigating potential gene functions. These CDM genes have a role in tumor cell survival and proliferation, suggesting that they may be used as new therapeutic targets for some malignant tumors. So far, there have been less research on the involvement of CDM genes in breast cancer, and only a tiny percentage of CDM genes have been studied. In this study, information of patients with breast cancer was extracted from The Cancer Genome Atlas (TCGA), from which differentially expressed CDM genes in breast cancer were determined. A variety of bioinformatics techniques were used to assess the functions and prognostic relevance of these confirmed CDM genes. In all, 290 CDM genes were found differentially expressed. Six CDM genes (SRF, RAD51, PMF1, EXOSC3, EXOC1, and TSEN54) were found to be associated with the prognosis of breast cancer samples. Based on the expression of the identified CDM genes and their coefficients, a prognosis model was constructed for prediction, according to which patients with breast cancer were separated into two risk groups. Those with high risk had substantially poorer overall survival (OS) than patients in the other risk group in the TCGA training set, TCGA testing set, and an external cohort from Gene Expression Omnibus (GEO) database. The area under the receiver operating characteristic (ROC) curve for this prognostic signature was, respectively, 0.717 and 0.635 for TCGA training and testing sets, demonstrating its reliability in predicting the prognosis of patients with breast cancer. We next created a nomogram using the six CDM genes discovered to create a therapeutically useful model. The Human Protein Atlas database was used to acquire all immunohistochemistry staining images of the discovered CDM genes. The proportions of tumor-infiltrating immune cells, as well as the expression levels of checkpoint genes, varied substantially between the two risk groups, according to the analyses of immune response. In conclusion, the findings of this research may aid in the understanding of the prognostic value and biological roles of CDM genes in breast cancer.
Collapse
|
3
|
Diehl JN, Klomp JE, Snare KR, Hibshman PS, Blake DR, Kaiser ZD, Gilbert TSK, Baldelli E, Pierobon M, Papke B, Yang R, Hodge RG, Rashid NU, Petricoin EF, Herring LE, Graves LM, Cox AD, Der CJ. The KRAS-regulated kinome identifies WEE1 and ERK coinhibition as a potential therapeutic strategy in KRAS-mutant pancreatic cancer. J Biol Chem 2021; 297:101335. [PMID: 34688654 PMCID: PMC8591367 DOI: 10.1016/j.jbc.2021.101335] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 09/24/2021] [Accepted: 10/19/2021] [Indexed: 02/07/2023] Open
Abstract
Oncogenic KRAS drives cancer growth by activating diverse signaling networks, not all of which have been fully delineated. We set out to establish a system-wide profile of the KRAS-regulated kinase signaling network (kinome) in KRAS-mutant pancreatic ductal adenocarcinoma (PDAC). We knocked down KRAS expression in a panel of six cell lines and then applied multiplexed inhibitor bead/MS to monitor changes in kinase activity and/or expression. We hypothesized that depletion of KRAS would result in downregulation of kinases required for KRAS-mediated transformation and in upregulation of other kinases that could potentially compensate for the deleterious consequences of the loss of KRAS. We identified 15 upregulated and 13 downregulated kinases in common across the panel of cell lines. In agreement with our hypothesis, all 15 of the upregulated kinases have established roles as cancer drivers (e.g., SRC, TGF-β1, ILK), and pharmacological inhibition of one of these upregulated kinases, DDR1, suppressed PDAC growth. Interestingly, 11 of the 13 downregulated kinases have established driver roles in cell cycle progression, particularly in mitosis (e.g., WEE1, Aurora A, PLK1). Consistent with a crucial role for the downregulated kinases in promoting KRAS-driven proliferation, we found that pharmacological inhibition of WEE1 also suppressed PDAC growth. The unexpected paradoxical activation of ERK upon WEE1 inhibition led us to inhibit both WEE1 and ERK concurrently, which caused further potent growth suppression and enhanced apoptotic death compared with WEE1 inhibition alone. We conclude that system-wide delineation of the KRAS-regulated kinome can identify potential therapeutic targets for KRAS-mutant pancreatic cancer.
Collapse
Affiliation(s)
- J Nathaniel Diehl
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jennifer E Klomp
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kayla R Snare
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Priya S Hibshman
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Cell Biology and Physiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Devon R Blake
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Zane D Kaiser
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Thomas S K Gilbert
- Cell Biology and Physiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; UNC Michael Hooker Proteomics Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Elisa Baldelli
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia, USA
| | - Mariaelena Pierobon
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia, USA
| | - Björn Papke
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Runying Yang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Richard G Hodge
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Naim U Rashid
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia, USA
| | - Laura E Herring
- Cell Biology and Physiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; UNC Michael Hooker Proteomics Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lee M Graves
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Cell Biology and Physiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; UNC Michael Hooker Proteomics Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Adrienne D Cox
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Cell Biology and Physiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Channing J Der
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Cell Biology and Physiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| |
Collapse
|
4
|
Lockhead S, Moskaleva A, Kamenz J, Chen Y, Kang M, Reddy AR, Santos SDM, Ferrell JE. The Apparent Requirement for Protein Synthesis during G2 Phase Is due to Checkpoint Activation. Cell Rep 2021; 32:107901. [PMID: 32668239 PMCID: PMC7802425 DOI: 10.1016/j.celrep.2020.107901] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 05/12/2020] [Accepted: 06/22/2020] [Indexed: 11/18/2022] Open
Abstract
Protein synthesis inhibitors (e.g., cycloheximide) block mitotic entry, suggesting that cell cycle progression requires protein synthesis until right before mitosis. However, cycloheximide is also known to activate p38 mitogen-activated protein kinase (MAPK), which can delay mitotic entry through a G2/M checkpoint. Here, we ask whether checkpoint activation or a requirement for protein synthesis is responsible for the cycloheximide effect. We find that p38 inhibitors prevent cycloheximide-treated cells from arresting in G2 phase and that G2 duration is normal in approximately half of these cells. The Wee1 inhibitor MK-1775 and Wee1/Myt1 inhibitor PD0166285 also prevent cycloheximide from blocking mitotic entry, raising the possibility that Wee1 and/or Myt1 mediate the cycloheximide-induced G2 arrest. Thus, protein synthesis during G2 phase is not required for mitotic entry, at least when the p38 checkpoint pathway is abrogated. However, M phase progression is delayed in cycloheximide-plus-kinase-inhibitor-treated cells, emphasizing the different requirements of protein synthesis for timely entry and completion of mitosis. Protein synthesis inhibitors have long been known to prevent G2 phase cells from entering mitosis. Lockhead et al. demonstrate that this G2 arrest is due to the activation of p38 MAPK, not insufficient protein synthesis, arguing that protein synthesis in G2 phase is not absolutely required for mitotic entry.
Collapse
Affiliation(s)
- Sarah Lockhead
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305-5174, USA
| | - Alisa Moskaleva
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305-5174, USA
| | - Julia Kamenz
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305-5174, USA.
| | - Yuxin Chen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305-5174, USA
| | - Minjung Kang
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305-5174, USA
| | - Anay R Reddy
- Department of Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Silvia D M Santos
- Quantitative Cell Biology Laboratory, The Francis Crick Institute, London, UK
| | - James E Ferrell
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305-5174, USA; Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305-5307, USA.
| |
Collapse
|
5
|
Boyle AJ, Cao PJ, Cai Z, Chan C, Hedley DW, Reilly RM. Radioimmunotherapy of human pancreatic cancer xenografts in NOD-scid mice with [ 64Cu]Cu-NOTA-panitumumab F(ab') 2 alone or combined with radiosensitizing gemcitabine and the PARP inhibitor, rucaparib. Nucl Med Biol 2020; 84-85:46-54. [PMID: 32062317 DOI: 10.1016/j.nucmedbio.2020.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/20/2020] [Accepted: 02/02/2020] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Our objective was to determine the feasibility of extending our previously reported PET imaging study of pancreatic cancer (PnCa) with [64Cu]Cu-NOTA-panitumumab F(ab')2 to radioimmunotherapy (RIT) by exploiting the β-particle and Auger electron emissions of 64Cu (PET theranostic concept). To enhance the effectiveness of [64Cu]Cu-NOTA-panitumumab F(ab')2, we further combined RIT with radiosensitizing gemcitabine (GEM) and the poly(ADP)ribose polymerase inhibitor (PARPi), rucaparib. METHODS Normal tissue toxicity was assessed in non-tumor-bearing NOD-scid mice injected i.v. with [64Cu]Cu-NOTA-panitumumab F(ab')2 (1.85-9.25 MBq; 10 μg) or [64Cu]Cu-NOTA-anti-mouse EGFR Ab30 F(ab')2 (12.95 MBq). Body weight was monitored, and hematopoietic (CBC), liver (ALT) and kidney [creatinine (SCr)] toxicity were assessed. RIT studies were performed in NOD-scid mice with s.c. OCIP23 human PnCa patient-derived xenografts (PDX) administered [64Cu]Cu-NOTA-panitumumab F(ab')2 (3.7 MBq; 10 μg), unlabeled panitumumab F(ab')2 (10 μg) or normal saline every two weeks. Subsequent studies evaluated RIT with [64Cu]Cu-NOTA-panitumumab F(ab')2 (12.95 MBq; 10 μg) administered alone or combined with GEM and the PARPi, rucaparib administered on a 14-day treatment cycle for up to 6 cycles in NOD-scid mice with s.c. PANC-1 human PnCa xenografts. The radiation absorbed dose in PANC-1 tumors and normal organs in mice after a single i.v. injection of [64Cu]Cu-NOTA-panitumumab F(ab')2 (12.95 MBq; 10 μg) was estimated based on previously reported biodistribution studies of [64Cu]Cu-NOTA-panitumumab F(ab')2. RESULTS No normal tissue toxicity was observed in non-tumor-bearing NOD-scid mice administered up to 3.7 MBq (10 μg) of [64Cu]Cu-NOTA-panitumumab F(ab')2 but slightly increased ALT was noted at 9.25 MBq. Administration of [64Cu]Cu-NOTA-anti-mouse EGFR Ab30 F(ab')2 (12.95 MBq; 10 μg) caused some hematopoietic toxicity but no increase in ALT or SCr or decreased body weight. A slight tumor growth delay and increased survival was noted in NOD-scid mice with s.c. OCIP23 PDX treated with [64Cu]Cu-NOTA-panitumumab F(ab')2 (3.7 MBq; 10 μg) or unlabeled panitumumab F(ab')2 (10 μg) compared to normal saline treated mice. RIT with [64Cu]Cu-NOTA-panitumumab F(ab')2 (12.95 MBq; 10 μg) combined with GEM + PARPi for up to 6 cycles was most effective for the treatment of PANC-1 tumors. Tumor doubling time increased to 13.3 ± 0.9 days vs. 7.8 ± 3.7 days for RIT alone and 9.3 ± 2.2 days for normal saline treatment. Median survival was significantly longer (P < 0.05) than in mice treated with normal saline (35 days) for RIT + GEM + PARPi (71 days), GEM + PARPi (44 days) and RIT + GEM (43 days) but not for RIT alone (25 days). RIT + GEM + PARPi provided a longer median survival than RIT (P < 0.01), GEM + PARPi (P = 0.01) but not RIT + GEM (P = 0.23). Nonetheless, PANC-1 tumors grew exponentially in all treatment groups. The absorbed dose in PANC-1 tumors after a single i.v. injection of [64Cu]Cu-NOTA-panitumumab F(ab')2 (12.85 MBq; 10 μg) was 0.8 Gy, while the dose in normal organs ranged from 0.6-1.2 Gy. CONCLUSIONS We conclude that RIT with [64Cu]Cu-NOTA-panitumumab F(ab')2 did not cause significant normal tissue toxicity but was not effective when administered alone for treatment of PnCa xenografts in NOD-scid mice. Combining RIT with GEM and the PARPi, rucaparib enhanced its effectiveness but tumors continued to grow exponentially. Our results suggest that 64Cu is not feasible for RIT of PnCa due to low tumor absorbed doses. 177Lu which has a higher abundance of moderate energy β-particle emissions may be more effective than 64Cu. The hematopoietic toxicity of [64Cu]Cu-NOTA-anti-mouse EGFR Ab30 F(ab')2 may be mediated by binding to mouse EGFR expressed on some hematopoietic stem cells. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE Direct extension of PET with 64Cu(Cu)-NOTA-panitumumab F(ab')2 to RIT exploiting the β-particle and Auger electron emissions of 64Cu is not feasible. Theranostic approaches that combine PET with RIT employing 177Lu may be more promising and should be explored.
Collapse
Affiliation(s)
- Amanda J Boyle
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Ping-Jiang Cao
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Zhongli Cai
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Conrad Chan
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - David W Hedley
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Raymond M Reilly
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada; Department of Medical Imaging, University of Toronto, Toronto, ON, Canada; Toronto General Research Institute, Joint Department of Medical Imaging, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
6
|
Dhahbi J, Nunez Lopez YO, Schneider A, Victoria B, Saccon T, Bharat K, McClatchey T, Atamna H, Scierski W, Golusinski P, Golusinski W, Masternak MM. Profiling of tRNA Halves and YRNA Fragments in Serum and Tissue From Oral Squamous Cell Carcinoma Patients Identify Key Role of 5' tRNA-Val-CAC-2-1 Half. Front Oncol 2019; 9:959. [PMID: 31616639 PMCID: PMC6775249 DOI: 10.3389/fonc.2019.00959] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common type of head and neck cancer and, as indicated by The Oral Cancer Foundation, kills at an alarming rate of roughly one person per hour. With this study, we aimed at better understanding disease mechanisms and identifying minimally invasive disease biomarkers by profiling novel small non-coding RNAs (specifically, tRNA halves and YRNA fragments) in both serum and tumor tissue from humans. Small RNA-Sequencing identified multiple 5' tRNA halves and 5' YRNA fragments that displayed significant differential expression levels in circulation and/or tumor tissue, as compared to control counterparts. In addition, by implementing a modification of weighted gene coexpression network analysis, we identified an upregulated genetic module comprised of 5' tRNA halves and miRNAs (miRNAs were described in previous study using the same samples) with significant association with the cancer trait. By consequently implementing miRNA-overtargeting network analysis, the biological function of the module (and by "guilt by association," the function of the 5' tRNA-Val-CAC-2-1 half) was found to involve the transcriptional targeting of specific genes involved in the negative regulation of the G1/S transition of the mitotic cell cycle. These findings suggest that 5' tRNA-Val-CAC-2-1 half (reduced in serum of OSCC patients and elevated in the tumor tissue) could potentially serve as an OSCC circulating biomarker and/or target for novel anticancer therapies. To our knowledge, this is the first time that the specific molecular function of a 5'-tRNA half is specifically pinpointed in OSCC.
Collapse
Affiliation(s)
- Joseph Dhahbi
- Department of Medical Education, School of Medicine, California University of Science & Medicine, San Bernardino, CA, United States
| | - Yury O. Nunez Lopez
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, FL, United States
| | - Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Berta Victoria
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Tatiana Saccon
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, Brazil
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Krish Bharat
- Department of Medical Education, School of Medicine, California University of Science & Medicine, San Bernardino, CA, United States
| | - Thaddeus McClatchey
- Department of Medical Education, School of Medicine, California University of Science & Medicine, San Bernardino, CA, United States
| | - Hani Atamna
- Department of Medical Education, School of Medicine, California University of Science & Medicine, San Bernardino, CA, United States
| | - Wojciech Scierski
- Department of Otorhinolaryngology and Laryngological Oncology in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Pawel Golusinski
- Department of Otolaryngology and Maxillofacial Surgery, University of Zielona Gora, Zielona Gora, Poland
- Department of Biology and Environmental Studies, Poznan University of Medical Sciences, Poznań, Poland
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, The Greater Poland Cancer Centre, Poznań, Poland
| | - Wojciech Golusinski
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, The Greater Poland Cancer Centre, Poznań, Poland
| | - Michal M. Masternak
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
- Department of Biology and Environmental Studies, Poznan University of Medical Sciences, Poznań, Poland
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, The Greater Poland Cancer Centre, Poznań, Poland
| |
Collapse
|
7
|
Wu M, Li X, Liu Q, Xie Y, Yuan J, Wanggou S. miR-526b-3p serves as a prognostic factor and regulates the proliferation, invasion, and migration of glioma through targeting WEE1. Cancer Manag Res 2019; 11:3099-3110. [PMID: 31114353 PMCID: PMC6489667 DOI: 10.2147/cmar.s192361] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/17/2019] [Indexed: 12/22/2022] Open
Abstract
Background: MicroRNAs play important roles in cancer progression including glioma. In this study, we aimed to explore the expression pattern, prognostic potential, and functional role of miR-526b-3p in human glioma. Materials and methods: The expression of miR-526b-3p in glioma tissues and the adjacent non-tumor tissues was determined by quantitative RT-PCR. The chi-square test was performed to evaluate the statistical associations between miR-526b-3p level and patient characteristics. The prognostic value of miR-526b-3p was analyzed by Kaplan–Meier and Cox regression analyses. The function of miR-526b-3p was analyzed by MTT, colony formation assay, transwell assay, and flow cytometry analysis in vitro. The binding between miR-526b-3p and predicted target WEE1 was verified using dual luciferase assay and Western blot analysis. Results: We found that miR-526b-3p expression was significantly downregulated in both glioma tissues and cell lines. Downregulation of miR-526b-3p was significantly associated with advanced WHO grade, lower KPS score, and inferior patient outcomes. Functional investigation indicated that overexpression of miR-526b-3p suppressed cell proliferation, migration, and invasion, and promoted apoptosis in glioma cell lines. Mechanically, WEE1 was identified as direct targets of miR-526b-3p and overexpression of WEE1 significantly suppressed the levels of WEE1. Moreover, re-introduction of WEE1 abrogates the suppression of motility and invasiveness induced by miR-526b-3p in glioma cells. Conclusion: These findings indicate that miR-526b-3p may target WEE1 and inhibit glioma tumorigenesis and progression.
Collapse
Affiliation(s)
- Ming Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Yuanyang Xie
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Jian Yuan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Siyi Wanggou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
8
|
Lin X, Chen D, Zhang C, Zhang X, Li Z, Dong B, Gao J, Shen L. Augmented antitumor activity by olaparib plus AZD1775 in gastric cancer through disrupting DNA damage repair pathways and DNA damage checkpoint. J Exp Clin Cancer Res 2018; 37:129. [PMID: 29954437 PMCID: PMC6027790 DOI: 10.1186/s13046-018-0790-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 06/13/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Targeting poly ADP-ribose polymerase (PARP) has been recently identified as a promising option against gastric cancer (GC). However, PARP inhibitors alone achieve limited efficacy. Combination strategies, especially with homologous recombination (HR) impairment, are of great hope to optimize PARP inhibitor's efficacy and expand target populations but remains largely unknown. Herein, we investigated whether a WEE1/ Polo-like kinase 1 (PLK1) dual inhibitor AZD1775 reported to impair HR augmented anticancer activity of a PARP inhibitor olaparib and its underlying mechanisms. METHODS GC cell lines and in vivo xenografts were employed to determine antitumor activity of PARP inhibitor combined with WEE1/PLK1 dual inhibitor AZD1775. Western blot, genetic knockdown by siRNA, flow cytometry, Immunohistochemistry were performed to explore the underlying mechanisms. RESULTS AZD1775 dually targeting WEE1/PLK1 enhanced effects of olaparib on growth inhibition and apoptotic induction in GC cells. Mechanistic investigations elucidate that WEE1/PLK1 blockade downregulated several HR-related proteins and caused an accumulation in γH2AX. As confirmed in both GC cell lines and mice bearing GC xenografts, these effects were enhanced by AZD1775-olaparib combination compared to olaparib alone, suggesting that disrupting HR-mediated DNA damage repairs (DDR) by WEE1/PLK1 blockade might be responsible for improved GC cells' response to PARP inhibitors. Given the DNA damage checkpoint as a primary target of WEE1 inhibition, our data also demonstrate that AZD1775 abrogated olaparib-activated DNA damage checkpoint through CDC2 de-phosphorylation, followed by mitotic progression with unrepaired DNA damage (marked by increased pHH3-stained and γH2AX-stained cells, respectively). CONCLUSIONS PARP inhibitor olaparib combined with WEE1/PLK1 dual inhibitor AZD1775 elicited potentiated anticancer activity through disrupting DDR signaling and the DNA damage checkpoint. It sheds light on the combination strategy of WEE1/PLK1 dual inhibitors with PARP inhibitors in the treatment of GC, even in HR-proficient patients.
Collapse
Affiliation(s)
- Xiaoting Lin
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142 China
| | - Dongshao Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142 China
| | - Cheng Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142 China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142 China
| | - Zhongwu Li
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142 China
| | - Bin Dong
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142 China
| | - Jing Gao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142 China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142 China
| |
Collapse
|
9
|
Wee1 Inhibitor AZD1775 Combined with Cisplatin Potentiates Anticancer Activity against Gastric Cancer by Increasing DNA Damage and Cell Apoptosis. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5813292. [PMID: 29977914 PMCID: PMC6011131 DOI: 10.1155/2018/5813292] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/06/2018] [Accepted: 05/10/2018] [Indexed: 12/30/2022]
Abstract
Based on the mechanisms by which Wee1 inhibitor and cisplatin played their own role, a promising strategy of Wee1 inhibitor combined with cisplatin was proposed, which was investigated in gastric cancer (GC). Either Wee1 inhibitor AZD1775 or cisplatin alone had a certain inhibitory effect on in vitro cell proliferation; however, the inhibitory effect was more significant when AZD1775 combined with cisplatin in vitro and in vivo. The underlying mechanisms unveiled that the increased DNA damage indicated by increased γH2AX protein, as well as augmented cell apoptosis indicated by upregulated proapoptotic proteins, was responsible for the significant inhibitory effect of AZD1775 plus cisplatin. Moreover, compared to any single drug, in vitro cell migration and invasion abilities were further attenuated by AZD1775 combined with cisplatin. There were suggestive results that the potentiated cytotoxicity between AZD1775 and cisplatin deserved a deep exploration in the future.
Collapse
|