1
|
Sellars E, Savguira M, Wu J, Cancelliere S, Jen M, Krishnan R, Hakem A, Barsyte-Lovejoy D, Hakem R, Narod SA, Kotsopoulos J, Salmena L. A high-throughput approach to identify BRCA1-downregulating compounds to enhance PARP inhibitor sensitivity. iScience 2024; 27:110180. [PMID: 38993666 PMCID: PMC11238136 DOI: 10.1016/j.isci.2024.110180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/29/2024] [Accepted: 06/01/2024] [Indexed: 07/13/2024] Open
Abstract
PARP inhibitors (PARPi) are efficacious in BRCA1-null tumors; however, their utility is limited in tumors with functional BRCA1. We hypothesized that pharmacologically reducing BRCA1 protein levels could enhance PARPi effectiveness in BRCA1 wild-type tumors. To identify BRCA1 downregulating agents, we generated reporter cell lines using CRISPR-mediated editing to tag endogenous BRCA1 protein with HiBiT. These reporter lines enable the sensitive measurement of BRCA1 protein levels by luminescence. Validated reporter cells were used in a pilot screen of epigenetic-modifying probes and a larger screen of more than 6,000 compounds. We identified 7 compounds that could downregulate BRCA1-HiBiT expression and synergize with olaparib. Three compounds, N-acetyl-N-acetoxy chlorobenzenesulfonamide (NANAC), A-443654, and CHIR-124, were validated to reduce BRCA1 protein levels and sensitize breast cancer cells to the toxic effects of olaparib. These results suggest that BRCA1-HiBiT reporter cells hold promise in developing agents to improve the clinical utility of PARPi.
Collapse
Affiliation(s)
- Erin Sellars
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Women's College Research Institute, Women's College Hospital, Toronto, ON M5S 1B2, Canada
| | - Margarita Savguira
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jie Wu
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sabrina Cancelliere
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Mark Jen
- Lunenfeld-Tanenbaum Research Institute, Network Biology Collaborative Centre, High-Throughput Screening, Mt. Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Rehna Krishnan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Anne Hakem
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Dalia Barsyte-Lovejoy
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Structural Genomics Consortium, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Razqallah Hakem
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Steven A Narod
- Women's College Research Institute, Women's College Hospital, Toronto, ON M5S 1B2, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Joanne Kotsopoulos
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Women's College Research Institute, Women's College Hospital, Toronto, ON M5S 1B2, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Leonardo Salmena
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Women's College Research Institute, Women's College Hospital, Toronto, ON M5S 1B2, Canada
| |
Collapse
|
2
|
Candido MF, Medeiros M, Veronez LC, Bastos D, Oliveira KL, Pezuk JA, Valera ET, Brassesco MS. Drugging Hijacked Kinase Pathways in Pediatric Oncology: Opportunities and Current Scenario. Pharmaceutics 2023; 15:pharmaceutics15020664. [PMID: 36839989 PMCID: PMC9966033 DOI: 10.3390/pharmaceutics15020664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Childhood cancer is considered rare, corresponding to ~3% of all malignant neoplasms in the human population. The World Health Organization (WHO) reports a universal occurrence of more than 15 cases per 100,000 inhabitants around the globe, and despite improvements in diagnosis, treatment and supportive care, one child dies of cancer every 3 min. Consequently, more efficient, selective and affordable therapeutics are still needed in order to improve outcomes and avoid long-term sequelae. Alterations in kinases' functionality is a trademark of cancer and the concept of exploiting them as drug targets has burgeoned in academia and in the pharmaceutical industry of the 21st century. Consequently, an increasing plethora of inhibitors has emerged. In the present study, the expression patterns of a selected group of kinases (including tyrosine receptors, members of the PI3K/AKT/mTOR and MAPK pathways, coordinators of cell cycle progression, and chromosome segregation) and their correlation with clinical outcomes in pediatric solid tumors were accessed through the R2: Genomics Analysis and Visualization Platform and by a thorough search of published literature. To further illustrate the importance of kinase dysregulation in the pathophysiology of pediatric cancer, we analyzed the vulnerability of different cancer cell lines against their inhibition through the Cancer Dependency Map portal, and performed a search for kinase-targeted compounds with approval and clinical applicability through the CanSAR knowledgebase. Finally, we provide a detailed literature review of a considerable set of small molecules that mitigate kinase activity under experimental testing and clinical trials for the treatment of pediatric tumors, while discuss critical challenges that must be overcome before translation into clinical options, including the absence of compounds designed specifically for childhood tumors which often show differential mutational burdens, intrinsic and acquired resistance, lack of selectivity and adverse effects on a growing organism.
Collapse
Affiliation(s)
- Marina Ferreira Candido
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Mariana Medeiros
- Regional Blood Center, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Luciana Chain Veronez
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - David Bastos
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Karla Laissa Oliveira
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Julia Alejandra Pezuk
- Departament of Biotechnology and Innovation, Anhanguera University of São Paulo, UNIAN/SP, São Paulo 04119-001, SP, Brazil
| | - Elvis Terci Valera
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - María Sol Brassesco
- Departament of Biotechnology and Innovation, Anhanguera University of São Paulo, UNIAN/SP, São Paulo 04119-001, SP, Brazil
- Correspondence: ; Tel.: +55-16-3315-9144; Fax: +55-16-3315-4886
| |
Collapse
|
3
|
He XL, Hu YH, Chen JM, Zhang DQ, Yang HL, Zhang LZ, Mu YP, Zhang H, Chen GF, Liu W, Liu P. SNS-032 attenuates liver fibrosis by anti-active hepatic stellate cells via inhibition of cyclin dependent kinase 9. Front Pharmacol 2022; 13:1016552. [PMID: 36313366 PMCID: PMC9597511 DOI: 10.3389/fphar.2022.1016552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
Liver fibrosis is a common pathological process of all chronic liver diseases. Hepatic stellate cells (HSCs) play a central role in the development of liver fibrosis. Cyclin-dependent kinase 9 (CDK9) is a cell cycle kinase that regulates mRNA transcription and elongation. A CDK9 inhibitor SNS-032 has been reported to have good effects in anti-tumor. However, the role of SNS-032 in the development of liver fibrosis is unclear. In this study, SNS-032 was found to alleviate hepatic fibrosis by inhibiting the activation and inducing the apoptosis of active HSCs in carbon tetrachloride-induced model mice. In vitro, SNS-032 inhibited the activation and proliferation of active HSCs and induced the apoptosis of active HSCs by downregulating the expression of CDK9 and its downstream signal transductors, such phosphorylated RNA polymerase II and Bcl-2. CDK9 short hairpin RNA was transfected into active HSCs to further elucidate the mechanism of the above effects. Similar results were observed in active HSCs after CDK9 knockdown. In active HSCs with CDK9 knockdown, the expression levels of CDK9, phosphorylated RNA polymerase II, XIAP, Bcl-2, Mcl-1, and ɑ-SMA significantly decreased, whereas those of cleaved-PARP1 and Bax decreased prominently. These results indicated that SNS-032 is a potential drug and CDK9 might be a new prospective target for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Xiao-Li He
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Endocrinology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-Hong Hu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jia-Mei Chen
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ding-Qi Zhang
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hai-Lin Yang
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lin-Zhang Zhang
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-Ping Mu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Zhang
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Gao-Feng Chen
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Liu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Wei Liu, ; Ping Liu,
| | - Ping Liu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Wei Liu, ; Ping Liu,
| |
Collapse
|
4
|
CDK Inhibition Primes for Anti-PD-L1 Treatment in Triple-Negative Breast Cancer Models. Cancers (Basel) 2022; 14:cancers14143361. [PMID: 35884422 PMCID: PMC9322647 DOI: 10.3390/cancers14143361] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/29/2022] [Accepted: 07/07/2022] [Indexed: 02/01/2023] Open
Abstract
Triple-negative breast cancers (TNBC) expressing PD-L1 qualify for checkpoint inhibitor immunotherapy. Cyclin E/CDK2 is a potential target axis in TNBC; however, small-molecule drugs at efficacious doses may be associated with toxicity, and treatment alongside immunotherapy requires investigation. We evaluated CDK inhibition at suboptimal levels and its anti-tumor and immunomodulatory effects. Transcriptomic analyses of primary breast cancers confirmed higher cyclin E/CDK2 expression in TNBC compared with non-TNBC. Out of the three CDK2-targeting inhibitors tested, the CDK 2, 7 and 9 inhibitor SNS-032 was the most potent in reducing TNBC cell viability and exerted cytotoxicity against all eight TNBC cell lines evaluated in vitro. Suboptimal SNS-032 dosing elevated cell surface PD-L1 expression in surviving TNBC cells. In mice engrafted with human immune cells and challenged with human MDA-MB-231 TNBC xenografts in mammary fat pads, suboptimal SNS-032 dosing partially restricted tumor growth, enhanced the tumor infiltration of human CD45+ immune cells and elevated cell surface PD-L1 expression in surviving cancer cells. In tumor-bearing mice engrafted with human immune cells, the anti-PD-L1 antibody avelumab, given sequentially following suboptimal SNS-032 dosing, reduced tumor growth compared with SNS-032 alone or with avelumab without prior SNS-032 priming. CDK inhibition at suboptimal doses promotes immune cell recruitment to tumors, PD-L1 expression by surviving TNBC cells and may complement immunotherapy.
Collapse
|
5
|
Targeting cyclin-dependent kinase 9 in cancer therapy. Acta Pharmacol Sin 2022; 43:1633-1645. [PMID: 34811514 PMCID: PMC9253122 DOI: 10.1038/s41401-021-00796-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Cyclin-dependent kinase (CDK) 9 associates mainly with cyclin T1 and forms the positive transcription elongation factor b (p-TEFb) complex responsible for transcriptional regulation. It has been shown that CDK9 modulates the expression and activity of oncogenes, such as MYC and murine double minute 4 (MDM4), and it also plays an important role in development and/or maintenance of the malignant cell phenotype. Malfunction of CDK9 is frequently observed in numerous cancers. Recent studies have highlighted the function of CDK9 through a variety of mechanisms in cancers, including the formation of new complexes and epigenetic alterations. Due to the importance of CDK9 activation in cancer cells, CDK9 inhibitors have emerged as promising candidates for cancer therapy. Natural product-derived and chemically synthesized CDK9 inhibitors are being examined in preclinical and clinical research. In this review, we summarize the current knowledge on the role of CDK9 in transcriptional regulation, epigenetic regulation, and different cellular factor interactions, focusing on new advances. We show the importance of CDK9 in mediating tumorigenesis and tumor progression. Then, we provide an overview of some CDK9 inhibitors supported by multiple oncologic preclinical and clinical investigations. Finally, we discuss the perspective and challenge of CDK9 modulation in cancer.
Collapse
|
6
|
Anticancer and Antiangiogenic Activities of Novel α-Mangostin Glycosides in Human Hepatocellular Carcinoma Cells via Downregulation of c-Met and HIF-1α. Int J Mol Sci 2020; 21:ijms21114043. [PMID: 32516967 PMCID: PMC7312821 DOI: 10.3390/ijms21114043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and is a leading cause of cancer-related death worldwide. Therefore, exploring effective anticancer agents and their modes of action is essential for the prevention and treatment of HCC. Glycosylation can significantly improve the physicochemical and biological properties of small molecules, such as high solubility, stability increase, and lower toxicity. In the present study, for the first time, we evaluated the anticancer and antiangiogenic activities of α-mangostin-3-O-β-D-2-deoxyglucopyranoside (Man-3DG) and α-mangostin 6-O-β-D-2-deoxyglucopyranoside (Man-6DG), glycosides of α-mangostin, against human HCC cells. Our results demonstrated that Man-3DG and Man-6DG significantly suppressed the growth of three different HCC cells (Hep3B, Huh7, and HepG2) as well as the migration of Hep3B cells. Furthermore, they induced cell cycle arrest in the G0/G1 phases and apoptotic cell death by regulating apoptosis-related proteins of mitochondria in Hep3B cells. Noticeably, Man-3DG and Man-6DG also caused autophagy, while co-treatment of the α-mangostin glycosides with an autophagy inhibitor 3-MA enhanced the inhibitory effect on Hep3B cell growth in comparison to single agent treatment. Moreover, Man-3DG and Man-6DG inhibited the c-Met signaling pathway that plays a critical role in the pathogenesis of HCC. Furthermore, the α-mangostin glycosides decreased Hep3B cell-induced angiogenesis in vitro through the downregulation of hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF). Notably, Man-6DG more effectively inhibited the growth, tumorsphere formation, and expression of cancer stemness regulators compared to α-mangostin and Man-3DG in 3D spheroid-cultured Hep3B cells. These findings suggest that the α-mangostin glycosides might be promising anticancer agents for HCC treatment with superior pharmacological properties than the parent molecule α-mangostin.
Collapse
|
7
|
Juric V, Murphy B. Cyclin-dependent kinase inhibitors in brain cancer: current state and future directions. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:48-62. [PMID: 35582046 PMCID: PMC9094053 DOI: 10.20517/cdr.2019.105] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/11/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022]
Abstract
Cyclin-dependent kinases (CDKs) are important regulatory enzymes in the normal physiological processes that drive cell-cycle transitions and regulate transcription. Virtually all cancers harbour genomic alterations that lead to the constitutive activation of CDKs, resulting in the proliferation of cancer cells. CDK inhibitors (CKIs) are currently in clinical use for the treatment of breast cancer, combined with endocrine therapy. In this review, we describe the potential of CKIs for the treatment of cancer with specific focus on glioblastoma (GBM), the most common and aggressive primary brain tumour in adults. Despite intense effort to combat GBM with surgery, radiation and temozolomide chemotherapy, the median survival for patients is 15 months and the majority of patients experience disease recurrence within 6-8 months of treatment onset. Novel therapeutic approaches are urgently needed for both newly diagnosed and recurrent GBM patients. In this review, we summarise the current preclinical and clinical findings emphasising that CKIs could represent an exciting novel approach for GBM treatment.
Collapse
Affiliation(s)
- Viktorija Juric
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin D02, Ireland
| | - Brona Murphy
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin D02, Ireland
| |
Collapse
|
8
|
Jiao Y, Preston S, Hofmann A, Taki A, Baell J, Chang BCH, Jabbar A, Gasser RB. A perspective on the discovery of selected compounds with anthelmintic activity against the barber's pole worm-Where to from here? ADVANCES IN PARASITOLOGY 2020; 108:1-45. [PMID: 32291083 DOI: 10.1016/bs.apar.2019.12.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Parasitic roundworms (nematodes) cause substantial morbidity and mortality in animals worldwide. Anthelmintic treatment is central to controlling these worms, but widespread resistance to most of the commercially available anthelmintics for veterinary and agricultural use is compromising control, such that there is an urgency to discover new and effective drugs. The purpose of this article is to review information on parasitic nematodes, the treatment and control of parasitic nematode infections and aspects of discovering new anthelmintics in the context of anthelmintic resistance problems, and then to discuss some progress that our group has made in identifying selected compounds with activity against nematodes. The focus of our recent work has been on discovering new chemical entities and known drugs with anthelmintic activities against Haemonchus contortus as well as other socioeconomically important parasitic nematodes for subsequent development. Using whole worm-based phenotypic assays, we have been screening compound collections obtained via product-development-partnerships and/or collaborators, and active compounds have been assessed for their potential as anthelmintic candidates. Following the screening of 15,333 chemicals from five distinct compound collections against H. contortus, we have discovered one new chemical entity (designated SN00797439), two human kinase inhibitors (SNS-032 and AG-1295), 14 tetrahydroquinoxaline analogues, one insecticide (tolfenpyrad) and two tolfenpyrad (pyrazole-5-carboxamide) derivatives (a-15 and a-17) with anthelmintic activity in vitro. Some of these 20 'hit' compounds have selectivity against H. contortus in vitro when compared to particular human cell lines. In our opinion, some of these compounds could represent starting points for 'lead' development. Accordingly, the next research steps to be pursued include: (i) chemical optimisation of representative chemicals via structure-activity relationship (SAR) evaluations; (ii) assessment of the breadth of spectrum of anthelmintic activity on a range of other parasitic nematodes, such as strongyloids, ascaridoids, enoplids and filarioids; (iii) detailed investigations of the absorption, distribution, metabolism, excretion and toxicity (ADMET) of optimised chemicals with broad nematocidal or nematostatic activity; and (iv) establishment of the modes of action of lead candidates.
Collapse
Affiliation(s)
- Yaqing Jiao
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Sarah Preston
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia; Faculty of Science and Technology, Federation University, Ballarat, VIC, Australia
| | - Andreas Hofmann
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Aya Taki
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Jonathan Baell
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Bill C H Chang
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Abdul Jabbar
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia.
| | - Robin B Gasser
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
9
|
Flores G, Everett JH, Boguslawski EA, Oswald BM, Madaj ZB, Beddows I, Dikalov S, Adams M, Klumpp-Thomas CA, Kitchen-Goosen SM, Martin SE, Caplen NJ, Helman LJ, Grohar PJ. CDK9 Blockade Exploits Context-dependent Transcriptional Changes to Improve Activity and Limit Toxicity of Mithramycin for Ewing Sarcoma. Mol Cancer Ther 2020; 19:1183-1196. [PMID: 32127464 DOI: 10.1158/1535-7163.mct-19-0775] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/17/2019] [Accepted: 02/19/2020] [Indexed: 11/16/2022]
Abstract
There is a need to develop novel approaches to improve the balance between efficacy and toxicity for transcription factor-targeted therapies. In this study, we exploit context-dependent differences in RNA polymerase II processivity as an approach to improve the activity and limit the toxicity of the EWS-FLI1-targeted small molecule, mithramycin, for Ewing sarcoma. The clinical activity of mithramycin for Ewing sarcoma is limited by off-target liver toxicity that restricts the serum concentration to levels insufficient to inhibit EWS-FLI1. In this study, we perform an siRNA screen of the druggable genome followed by a matrix drug screen to identify mithramycin potentiators and a synergistic "class" effect with cyclin-dependent kinase 9 (CDK9) inhibitors. These CDK9 inhibitors enhanced the mithramycin-mediated suppression of the EWS-FLI1 transcriptional program leading to a shift in the IC50 and striking regressions of Ewing sarcoma xenografts. To determine whether these compounds may also be liver protective, we performed a qPCR screen of all known liver toxicity genes in HepG2 cells to identify mithramycin-driven transcriptional changes that contribute to the liver toxicity. Mithramycin induces expression of the BTG2 gene in HepG2 but not Ewing sarcoma cells, which leads to a liver-specific accumulation of reactive oxygen species (ROS). siRNA silencing of BTG2 rescues the induction of ROS and the cytotoxicity of mithramycin in these cells. Furthermore, CDK9 inhibition blocked the induction of BTG2 to limit cytotoxicity in HepG2, but not Ewing sarcoma cells. These studies provide the basis for a synergistic and less toxic EWS-FLI1-targeted combination therapy for Ewing sarcoma.
Collapse
Affiliation(s)
- Guillermo Flores
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan.,College of Human Medicine, Michigan State University, Grand Rapids, Michigan
| | - Joel H Everett
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| | - Elissa A Boguslawski
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Brandon M Oswald
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Zachary B Madaj
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, Grand Rapids, Michigan
| | - Ian Beddows
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, Grand Rapids, Michigan
| | - Sergey Dikalov
- The Free Radicals in Medicine Core, Division of Clinical Pharmacology Vanderbilt University Medical Center, Nashville, Tennessee
| | - Marie Adams
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, Grand Rapids, Michigan
| | - Carleen A Klumpp-Thomas
- Trans-NIH RNAi Screening Facility, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, Maryland
| | - Susan M Kitchen-Goosen
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Scott E Martin
- Trans-NIH RNAi Screening Facility, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, Maryland
| | - Natasha J Caplen
- Genetics Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Lee J Helman
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Patrick J Grohar
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan. .,Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland.,Department of Pediatrics, Vanderbilt University, Nashville, Tennessee.,Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, Michigan.,Division of Pediatric Hematology-Oncology, Helen DeVos Children's Hospital, Grand Rapids, Michigan.,Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
10
|
Löschmann N, Michaelis M, Rothweiler F, Voges Y, Balónová B, Blight BA, Cinatl J. ABCB1 as predominant resistance mechanism in cells with acquired SNS-032 resistance. Oncotarget 2018; 7:58051-58064. [PMID: 27517323 PMCID: PMC5295411 DOI: 10.18632/oncotarget.11160] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 07/27/2016] [Indexed: 12/31/2022] Open
Abstract
The CDK inhibitor SNS-032 had previously exerted promising anti-neuroblastoma activity via CDK7 and 9 inhibition. ABCB1 expression was identified as major determinant of SNS-032 resistance. Here, we investigated the role of ABCB1 in acquired SNS-032 resistance. In contrast to ABCB1-expressing UKF-NB-3 sub-lines resistant to other ABCB1 substrates, SNS-032-adapted UKF-NB-3 (UKF-NB-3rSNS- 032300nM) cells remained sensitive to the non-ABCB1 substrate cisplatin and were completely re-sensitized to cytotoxic ABCB1 substrates by ABCB1 inhibition. Moreover, UKF-NB-3rSNS-032300nM cells remained similarly sensitive to CDK7 and 9 inhibition as UKF-NB-3 cells. In contrast, SHEPrSNS-0322000nM, the SNS-032-resistant sub-line of the neuroblastoma cell line SHEP, displayed low level SNS-032 resistance also when ABCB1 was inhibited. This discrepancy may be explained by the higher SNS-032 concentrations that were used to establish SHEPrSNS-0322000nM cells, since SHEP cells intrinsically express ABCB1 and are less sensitive to SNS-032 (IC50 912 nM) than UKF-NB-3 cells (IC50 153 nM). In conclusion, we show that ABCB1 expression represents the primary (sometimes exclusive) resistance mechanism in neuroblastoma cells with acquired resistance to SNS-032. Thus, ABCB1 inhibitors may increase the SNS-032 efficacy in ABCB1-expressing cells and prolong or avoid resistance formation.
Collapse
Affiliation(s)
- Nadine Löschmann
- Institut für Medizinische Virologie, Klinikum der Goethe-Universität, 60596 Frankfurt am Main, Germany
| | - Martin Michaelis
- Centre for Molecular Processing and School of Biosciences, University of Kent, Canterbury, UK
| | - Florian Rothweiler
- Institut für Medizinische Virologie, Klinikum der Goethe-Universität, 60596 Frankfurt am Main, Germany
| | - Yvonne Voges
- Institut für Medizinische Virologie, Klinikum der Goethe-Universität, 60596 Frankfurt am Main, Germany
| | | | - Barry A Blight
- School of Physical Sciences, University of Kent, Canterbury, UK
| | - Jindrich Cinatl
- Institut für Medizinische Virologie, Klinikum der Goethe-Universität, 60596 Frankfurt am Main, Germany
| |
Collapse
|
11
|
Castro-Gamero AM, Pezuk JA, Brassesco MS, Tone LG. G2/M inhibitors as pharmacotherapeutic opportunities for glioblastoma: the old, the new, and the future. Cancer Biol Med 2018; 15:354-374. [PMID: 30766748 PMCID: PMC6372908 DOI: 10.20892/j.issn.2095-3941.2018.0030] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Glioblastoma (GBM) is one of the deadliest tumors and has a median survival of 3 months if left untreated. Despite advances in rationally targeted pharmacological approaches, the clinical care of GBM remains palliative in intent. Since the majority of altered signaling cascades involved in cancer establishment and progression eventually affect cell cycle progression, an alternative approach for cancer therapy is to develop innovative compounds that block the activity of crucial molecules needed by tumor cells to complete cell division. In this context, we review promising ongoing and future strategies for GBM therapeutics aimed towards G2/M inhibition such as anti-microtubule agents and targeted therapy against G2/M regulators like cyclin-dependent kinases, Aurora inhibitors, PLK1, BUB, 1, and BUBR1, and survivin. Moreover, we also include investigational agents in the preclinical and early clinical settings. Although several drugs were shown to be gliotoxic, most of them have not yet entered therapeutic trials. The use of either single exposure or a combination with novel compounds may lead to treatment alternatives for GBM patients in the near future.
Collapse
Affiliation(s)
- Angel Mauricio Castro-Gamero
- Human Genetics Laboratory, Institute of Natural Sciences, Federal University of Alfenas (UNIFAL-MG), Alfenas 37130-001, Brazil
| | - Julia Alejandra Pezuk
- Biotechnology and Innovation in Health Program and Pharmacy Program, Anhanguera University São Paulo (UNIAN-SP), São Paulo 05145-200, Brazil
| | - María Sol Brassesco
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, Brazil
| | - Luiz Gonzaga Tone
- Department of Pediatrics.,Department of Genetics, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| |
Collapse
|
12
|
Jiao Y, Preston S, Koehler AV, Stroehlein AJ, Chang BCH, Simpson KJ, Cowley KJ, Palmer MJ, Laleu B, Wells TNC, Jabbar A, Gasser RB. Screening of the 'Stasis Box' identifies two kinase inhibitors under pharmaceutical development with activity against Haemonchus contortus. Parasit Vectors 2017; 10:323. [PMID: 28679424 PMCID: PMC5499055 DOI: 10.1186/s13071-017-2246-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 06/13/2017] [Indexed: 01/06/2023] Open
Abstract
Background In partnership with the Medicines for Malaria Venture (MMV), we screened a collection (‘Stasis Box’) of 400 compounds (which have been in clinical development but have not been approved for illnesses other than neglected infectious diseases) for inhibitory activity against Haemonchus contortus, in order to attempt to repurpose some of the compounds to parasitic nematodes. Methods We assessed the inhibition of compounds on the motility and/or development of exsheathed third-stage (xL3s) and fourth-stage (L4) larvae of H. contortus using a whole-organism screening assay. Results In the primary screen, we identified compound MMV690767 (also known as SNS-032) that inhibited xL3 motility by ~70% at a concentration of 20 μM after 72 h as well as compound MMV079840 (also known as AG-1295), which induced a coiled xL3 phenotype, with ~50% inhibition on xL3 motility. Subsequently, we showed that SNS-032 (IC50 = 12.4 μM) and AG-1295 (IC50 = 9.92 ± 1.86 μM) had a similar potency to inhibit xL3 motility. Although neither SNS-032 nor AG-1295 had a detectable inhibitory activity on L4 motility, both compounds inhibited L4 development (IC50 values = 41.24 μM and 7.75 ± 0.94 μM for SNS-032 and AG-1295, respectively). The assessment of the two compounds for toxic effects on normal human breast epithelial (MCF10A) cells revealed that AG-1295 had limited cytotoxicity (IC50 > 100 μM), whereas SNS-032 was quite toxic to the epithelial cells (IC50 = 1.27 μM). Conclusions Although the two kinase inhibitors, SNS-032 and AG-1295, had moderate inhibitory activity on the motility or development of xL3s or L4s of H. contortus in vitro, further work needs to be undertaken to chemically alter these entities to achieve the potency and selectivity required for them to become nematocidal or nematostatic candidates. Electronic supplementary material The online version of this article (doi:10.1186/s13071-017-2246-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yaqing Jiao
- Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary School, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Sarah Preston
- Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary School, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Anson V Koehler
- Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary School, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Andreas J Stroehlein
- Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary School, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Bill C H Chang
- Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary School, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Kaylene J Simpson
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Parkville, VIC, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Karla J Cowley
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Michael J Palmer
- Medicines for Malaria Venture (MMV), Route de Pré-Bois 20, CH-1215, Geneva, Switzerland
| | - Benoît Laleu
- Medicines for Malaria Venture (MMV), Route de Pré-Bois 20, CH-1215, Geneva, Switzerland
| | - Timothy N C Wells
- Medicines for Malaria Venture (MMV), Route de Pré-Bois 20, CH-1215, Geneva, Switzerland
| | - Abdul Jabbar
- Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary School, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Robin B Gasser
- Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary School, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
13
|
Rahaman MH, Kumarasiri M, Mekonnen LB, Yu M, Diab S, Albrecht H, Milne RW, Wang S. Targeting CDK9: a promising therapeutic opportunity in prostate cancer. Endocr Relat Cancer 2016; 23:T211-T226. [PMID: 27582311 DOI: 10.1530/erc-16-0299] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 08/31/2016] [Indexed: 12/18/2022]
Abstract
Cyclin-dependent kinase 9 (CDK9) is a key transcriptional regulator and a lucrative target for cancer treatment. Targeting CDK9 can effectively confine the hyperactivity of androgen receptor and the constitutive expression of anti-apoptotic proteins; both being main causes of prostate cancer (PCa) development and progression. In castrate-resistant PCa, traditional therapies that only target androgen receptor (AR) have become obsolete due to reprograming in AR activity to make the cells independent of androgen. CDK9 inhibitors may provide a new and better therapeutic opportunity over traditional treatment options by targeting both androgen receptor activity and anti-apoptotic proteins, improving the chances of positive outcomes, especially in patients with the advanced disease. This review focuses on biological functions of CDK9, its involvement with AR and the potential for therapeutic opportunities in PCa treatment.
Collapse
Affiliation(s)
| | | | - Laychiluh B Mekonnen
- Centre for Drug Discovery and DevelopmentSansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Mingfeng Yu
- Centre for Drug Discovery and DevelopmentSansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Sarah Diab
- Centre for Drug Discovery and DevelopmentSansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Hugo Albrecht
- Centre for Drug Discovery and DevelopmentSansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Robert W Milne
- Centre for Drug Discovery and DevelopmentSansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Shudong Wang
- Centre for Drug Discovery and DevelopmentSansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
14
|
Pfankuche VM, Spitzbarth I, Lapp S, Ulrich R, Deschl U, Kalkuhl A, Baumgärtner W, Puff C. Reduced angiogenic gene expression in morbillivirus-triggered oncolysis in a translational model for histiocytic sarcoma. J Cell Mol Med 2016; 21:816-830. [PMID: 27860224 PMCID: PMC5345635 DOI: 10.1111/jcmm.13023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 09/30/2016] [Indexed: 12/19/2022] Open
Abstract
Histiocytic sarcoma represents a rare malignant tumour with a short survival time, indicating the need of novel treatment strategies including oncolytic virotherapy. The underlying molecular mechanisms of viral oncolysis are largely unknown. As cancer in companion animals shares striking similarities with human counterparts, we chose a permanent canine histiocytic sarcoma cell line (DH82 cells) to identify global transcriptome changes following infection with canine distemper virus (CDV), a paramyxovirus closely related to human measles virus. Microarray analysis identified 3054 differentially expressed probe sets (DEPs), encoding for 892 up‐ and 869 down‐regulated unique canine genes, respectively, in DH82 cells persistently infected with the vaccine strain Onderstepoort of CDV (DH82‐Ond‐pi), compared to non‐infected DH82 cells. Up‐regulated genes were predominantly related to immune processes, as demonstrated by functional enrichment analysis. Moreover, there was substantial enrichment of genes characteristic for classically activated M1 and alternatively activated M2 macrophages in DH82‐Ond‐pi; however, significant polarization into either of both categories was lacking. ‘Angiogenesis’ was the dominant enriched functional term for the down‐regulated genes, highlighting decreased blood vessel generation as a potential mechanism of paramyxovirus‐induced oncolysis in DH82 cells. The anti‐angiogenic effect of infection was verified by immunohistochemistry, which revealed a lower blood vessel density in an in vivo mouse model, xenotransplanted with DH82‐Ond‐pi, compared to mice transplanted with non‐infected DH82 cells. Reduction in angiogenesis appears to be an important oncolytic mechanism of CDV in DH82 cells, suggesting that similar mechanisms might account for human histiocytic sarcoma and maybe other tumours in conjunction with measles virus.
Collapse
Affiliation(s)
| | - Ingo Spitzbarth
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| | - Stefanie Lapp
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| | - Reiner Ulrich
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| | - Ulrich Deschl
- Department of Non-Clinical Drug Safety, Boehringer Ingelheim Pharma GmbH&Co KG, Biberach (Riß), Germany
| | - Arno Kalkuhl
- Department of Non-Clinical Drug Safety, Boehringer Ingelheim Pharma GmbH&Co KG, Biberach (Riß), Germany
| | | | - Christina Puff
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| |
Collapse
|
15
|
Han JM, Kwon HJ, Jung HJ. Tricin, 4',5,7-trihydroxy-3',5'-dimethoxyflavone, exhibits potent antiangiogenic activity in vitro. Int J Oncol 2016; 49:1497-504. [PMID: 27498749 DOI: 10.3892/ijo.2016.3645] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/22/2016] [Indexed: 11/05/2022] Open
Abstract
Tumor growth and metastasis depend on angiogenesis triggered by chemical signals, such as vascular endothelial growth factor (VEGF), released from tumor cells. Therefore, the specific perturbation of angiogenesis has been considered a powerful strategy for the treatment of cancer. Herein, we report that tricin, 4',5,7-trihydroxy-3',5'-dimethoxyflavone, exhibits potent antiangiogenic activity in vitro. Tricin effectively suppressed the proliferation as well as VEGF-induced invasion and tube formation of human umbilical vein endothelial cells (HUVECs) at subtoxic doses. Furthermore, tricin significantly inhibited the angiogenesis of the chorioallantoic membrane from growing chick embryos without showing cytotoxicity. We also found that tricin blocked tumor cell-induced angiogenesis. Notably, tricin downregulated not only the VEGFR2 signal transduction by reducing reactive oxygen species (ROS) generation in endothelial cells, but also the expression of VEGF by inhibiting hypoxia inducible factor-1α (HIF-1α) accumulation in tumor cells. Moreover, combined treatment with tricin and bevacizumab, an anti-VEGF drug, ameliorated the antiangiogenic effect of bevacizumab. Taken together, our findings demonstrate for the first time that tricin possesses promising antiangiogenic potential and thus may be applied to anticancer therapy by targeting tumor angiogenesis.
Collapse
Affiliation(s)
- Jang Mi Han
- Department of BT-Convergent Pharmaceutical Engineering, Sun Moon University, Chungnam 336-708, Republic of Korea
| | - Ho Jeong Kwon
- Department of Biotechnology, Yonsei University, Seodaemun-gu, Seoul 120-749, Republic of Korea
| | - Hye Jin Jung
- Department of BT-Convergent Pharmaceutical Engineering, Sun Moon University, Chungnam 336-708, Republic of Korea
| |
Collapse
|
16
|
Sánchez-Peris M, Falomir E, Murga J, Carda M, Marco JA. Synthesis and evaluation of biphenyl derivatives as potential downregulators of VEGF protein secretion and telomerase-related gene expressions. Bioorg Med Chem 2016; 24:3108-15. [DOI: 10.1016/j.bmc.2016.05.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 04/27/2016] [Accepted: 05/17/2016] [Indexed: 12/13/2022]
|
17
|
Řezníčková E, Weitensteiner S, Havlíček L, Jorda R, Gucký T, Berka K, Bazgier V, Zahler S, Kryštof V, Strnad M. Characterization of a Pyrazolo[4,3-d]pyrimidine Inhibitor of Cyclin-Dependent Kinases 2 and 5 and Aurora A With Pro-Apoptotic and Anti-Angiogenic ActivityIn Vitro. Chem Biol Drug Des 2015. [DOI: 10.1111/cbdd.12618] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Eva Řezníčková
- Laboratory of Growth Regulators & Department of Chemical Biology and Genetics; Centre of the Region Haná for Biotechnological and Agricultural Research; Palacký University and Institute of Experimental Botany AS CR; Šlechtitelů 27 78371 Olomouc Czech Republic
| | - Sabine Weitensteiner
- Department of Pharmacy; LMU Munich - Center for Drug Research - Pharmaceutical Biology; Butenandtstr. 5-13 81377 Munich Germany
| | - Libor Havlíček
- Isotope Laboratory; Institute of Experimental Botany ASCR; Vídeňská 1083 14220 Prague Czech Republic
| | - Radek Jorda
- Laboratory of Growth Regulators & Department of Chemical Biology and Genetics; Centre of the Region Haná for Biotechnological and Agricultural Research; Palacký University and Institute of Experimental Botany AS CR; Šlechtitelů 27 78371 Olomouc Czech Republic
| | - Tomáš Gucký
- Laboratory of Growth Regulators & Department of Chemical Biology and Genetics; Centre of the Region Haná for Biotechnological and Agricultural Research; Palacký University and Institute of Experimental Botany AS CR; Šlechtitelů 27 78371 Olomouc Czech Republic
| | - Karel Berka
- Regional Centre of Advanced Technologies and Materials; Department of Physical Chemistry; Faculty of Science; Palacký University; 17. listopadu 12 77146 Olomouc Czech Republic
| | - Václav Bazgier
- Laboratory of Growth Regulators & Department of Chemical Biology and Genetics; Centre of the Region Haná for Biotechnological and Agricultural Research; Palacký University and Institute of Experimental Botany AS CR; Šlechtitelů 27 78371 Olomouc Czech Republic
- Department of Physical Chemistry; Faculty of Science; Palacký University; 17. listopadu 1192/12 771 46 Olomouc Czech Republic
| | - Stefan Zahler
- Department of Pharmacy; LMU Munich - Center for Drug Research - Pharmaceutical Biology; Butenandtstr. 5-13 81377 Munich Germany
| | - Vladimír Kryštof
- Laboratory of Growth Regulators & Department of Chemical Biology and Genetics; Centre of the Region Haná for Biotechnological and Agricultural Research; Palacký University and Institute of Experimental Botany AS CR; Šlechtitelů 27 78371 Olomouc Czech Republic
| | - Miroslav Strnad
- Laboratory of Growth Regulators & Department of Chemical Biology and Genetics; Centre of the Region Haná for Biotechnological and Agricultural Research; Palacký University and Institute of Experimental Botany AS CR; Šlechtitelů 27 78371 Olomouc Czech Republic
| |
Collapse
|
18
|
Jarry M, Lecointre C, Malleval C, Desrues L, Schouft MT, Lejoncour V, Liger F, Lyvinec G, Joseph B, Loaëc N, Meijer L, Honnorat J, Gandolfo P, Castel H. Impact of meriolins, a new class of cyclin-dependent kinase inhibitors, on malignant glioma proliferation and neo-angiogenesis. Neuro Oncol 2014; 16:1484-98. [PMID: 24891448 DOI: 10.1093/neuonc/nou102] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Glioblastomas are the most frequent and most aggressive primary brain tumors in adults. The median overall survival is limited to a few months despite surgery, radiotherapy, and chemotherapy. It is now clearly established that hyperactivity of cyclin-dependent kinases (CDKs) is one of the processes underlying hyperproliferation and tumoral growth. The marine natural products meridianins and variolins, characterized as CDK inhibitors, display a kinase-inhibitory activity associated with cytotoxic effects. In order to improve selectivity and efficiency of these CDK inhibitors, a series of hybrid compounds called meriolins have been synthesized. METHODS The potential antitumoral activity of meriolins was investigated in vitro on glioma cell lines (SW1088 and U87), native neural cells, and a human endothelial cell line (HUV-EC-C). The impact of intraperitoneal or intratumoral administrations of meriolin 15 was evaluated in vivo on 2 different nude mice-xenografted glioma models. RESULTS Meriolins 3, 5, and 15 exhibited antiproliferative properties with nanomolar IC50 and induced cell-cycle arrest and CDK inhibition associated with apoptotic events in human glioma cell lines. These meriolins blocked the proliferation rate of HUV-EC-C through cell cycle arrest and apoptosis. In vivo, meriolin 15 provoked a robust reduction in tumor volume in spite of toxicity for highest doses, associated with inhibition of cell division, activation of caspase 3, reduction of CD133 cells, and modifications of the vascular architecture. CONCLUSION Meriolins, and meriolin 15 in particular, exhibit antiproliferative and proapoptotic activities on both glioma and intratumoral endothelial cells, constituting key promising therapeutic lead compounds for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Marie Jarry
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, Biomedical Research Institute (IRIB), PRES Normandy, TC2N network, University of Rouen, Mont-Saint-Aignan, France (M.J., C.L., L.D., M.-T.S., V.L., P.G., H.C.); Neuro-oncology department, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France (C.M., J.H.); Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292, Lyon, France (C.M., J.H.); University of Claude Bernard - Lyon 1, Villeurbanne, France (C.M., J.H.); Institut de Chimie et Biochimie Moléculaires et Supramoléculaires UMR 5246, University of Claude Bernard - Lyon 1, Villeurbanne, France (F.L., G.L., B.J., N.L.); Protein Phosphorylation & Human Disease Group & USR3151, Station Biologique, Roscoff, France (N.L., L.M.); ManRos Therapeutics, Roscoff, France (L.M.)
| | - Céline Lecointre
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, Biomedical Research Institute (IRIB), PRES Normandy, TC2N network, University of Rouen, Mont-Saint-Aignan, France (M.J., C.L., L.D., M.-T.S., V.L., P.G., H.C.); Neuro-oncology department, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France (C.M., J.H.); Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292, Lyon, France (C.M., J.H.); University of Claude Bernard - Lyon 1, Villeurbanne, France (C.M., J.H.); Institut de Chimie et Biochimie Moléculaires et Supramoléculaires UMR 5246, University of Claude Bernard - Lyon 1, Villeurbanne, France (F.L., G.L., B.J., N.L.); Protein Phosphorylation & Human Disease Group & USR3151, Station Biologique, Roscoff, France (N.L., L.M.); ManRos Therapeutics, Roscoff, France (L.M.)
| | - Céline Malleval
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, Biomedical Research Institute (IRIB), PRES Normandy, TC2N network, University of Rouen, Mont-Saint-Aignan, France (M.J., C.L., L.D., M.-T.S., V.L., P.G., H.C.); Neuro-oncology department, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France (C.M., J.H.); Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292, Lyon, France (C.M., J.H.); University of Claude Bernard - Lyon 1, Villeurbanne, France (C.M., J.H.); Institut de Chimie et Biochimie Moléculaires et Supramoléculaires UMR 5246, University of Claude Bernard - Lyon 1, Villeurbanne, France (F.L., G.L., B.J., N.L.); Protein Phosphorylation & Human Disease Group & USR3151, Station Biologique, Roscoff, France (N.L., L.M.); ManRos Therapeutics, Roscoff, France (L.M.)
| | - Laurence Desrues
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, Biomedical Research Institute (IRIB), PRES Normandy, TC2N network, University of Rouen, Mont-Saint-Aignan, France (M.J., C.L., L.D., M.-T.S., V.L., P.G., H.C.); Neuro-oncology department, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France (C.M., J.H.); Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292, Lyon, France (C.M., J.H.); University of Claude Bernard - Lyon 1, Villeurbanne, France (C.M., J.H.); Institut de Chimie et Biochimie Moléculaires et Supramoléculaires UMR 5246, University of Claude Bernard - Lyon 1, Villeurbanne, France (F.L., G.L., B.J., N.L.); Protein Phosphorylation & Human Disease Group & USR3151, Station Biologique, Roscoff, France (N.L., L.M.); ManRos Therapeutics, Roscoff, France (L.M.)
| | - Marie-Thérèse Schouft
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, Biomedical Research Institute (IRIB), PRES Normandy, TC2N network, University of Rouen, Mont-Saint-Aignan, France (M.J., C.L., L.D., M.-T.S., V.L., P.G., H.C.); Neuro-oncology department, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France (C.M., J.H.); Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292, Lyon, France (C.M., J.H.); University of Claude Bernard - Lyon 1, Villeurbanne, France (C.M., J.H.); Institut de Chimie et Biochimie Moléculaires et Supramoléculaires UMR 5246, University of Claude Bernard - Lyon 1, Villeurbanne, France (F.L., G.L., B.J., N.L.); Protein Phosphorylation & Human Disease Group & USR3151, Station Biologique, Roscoff, France (N.L., L.M.); ManRos Therapeutics, Roscoff, France (L.M.)
| | - Vadim Lejoncour
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, Biomedical Research Institute (IRIB), PRES Normandy, TC2N network, University of Rouen, Mont-Saint-Aignan, France (M.J., C.L., L.D., M.-T.S., V.L., P.G., H.C.); Neuro-oncology department, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France (C.M., J.H.); Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292, Lyon, France (C.M., J.H.); University of Claude Bernard - Lyon 1, Villeurbanne, France (C.M., J.H.); Institut de Chimie et Biochimie Moléculaires et Supramoléculaires UMR 5246, University of Claude Bernard - Lyon 1, Villeurbanne, France (F.L., G.L., B.J., N.L.); Protein Phosphorylation & Human Disease Group & USR3151, Station Biologique, Roscoff, France (N.L., L.M.); ManRos Therapeutics, Roscoff, France (L.M.)
| | - François Liger
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, Biomedical Research Institute (IRIB), PRES Normandy, TC2N network, University of Rouen, Mont-Saint-Aignan, France (M.J., C.L., L.D., M.-T.S., V.L., P.G., H.C.); Neuro-oncology department, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France (C.M., J.H.); Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292, Lyon, France (C.M., J.H.); University of Claude Bernard - Lyon 1, Villeurbanne, France (C.M., J.H.); Institut de Chimie et Biochimie Moléculaires et Supramoléculaires UMR 5246, University of Claude Bernard - Lyon 1, Villeurbanne, France (F.L., G.L., B.J., N.L.); Protein Phosphorylation & Human Disease Group & USR3151, Station Biologique, Roscoff, France (N.L., L.M.); ManRos Therapeutics, Roscoff, France (L.M.)
| | - Gildas Lyvinec
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, Biomedical Research Institute (IRIB), PRES Normandy, TC2N network, University of Rouen, Mont-Saint-Aignan, France (M.J., C.L., L.D., M.-T.S., V.L., P.G., H.C.); Neuro-oncology department, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France (C.M., J.H.); Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292, Lyon, France (C.M., J.H.); University of Claude Bernard - Lyon 1, Villeurbanne, France (C.M., J.H.); Institut de Chimie et Biochimie Moléculaires et Supramoléculaires UMR 5246, University of Claude Bernard - Lyon 1, Villeurbanne, France (F.L., G.L., B.J., N.L.); Protein Phosphorylation & Human Disease Group & USR3151, Station Biologique, Roscoff, France (N.L., L.M.); ManRos Therapeutics, Roscoff, France (L.M.)
| | - Benoît Joseph
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, Biomedical Research Institute (IRIB), PRES Normandy, TC2N network, University of Rouen, Mont-Saint-Aignan, France (M.J., C.L., L.D., M.-T.S., V.L., P.G., H.C.); Neuro-oncology department, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France (C.M., J.H.); Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292, Lyon, France (C.M., J.H.); University of Claude Bernard - Lyon 1, Villeurbanne, France (C.M., J.H.); Institut de Chimie et Biochimie Moléculaires et Supramoléculaires UMR 5246, University of Claude Bernard - Lyon 1, Villeurbanne, France (F.L., G.L., B.J., N.L.); Protein Phosphorylation & Human Disease Group & USR3151, Station Biologique, Roscoff, France (N.L., L.M.); ManRos Therapeutics, Roscoff, France (L.M.)
| | - Nadège Loaëc
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, Biomedical Research Institute (IRIB), PRES Normandy, TC2N network, University of Rouen, Mont-Saint-Aignan, France (M.J., C.L., L.D., M.-T.S., V.L., P.G., H.C.); Neuro-oncology department, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France (C.M., J.H.); Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292, Lyon, France (C.M., J.H.); University of Claude Bernard - Lyon 1, Villeurbanne, France (C.M., J.H.); Institut de Chimie et Biochimie Moléculaires et Supramoléculaires UMR 5246, University of Claude Bernard - Lyon 1, Villeurbanne, France (F.L., G.L., B.J., N.L.); Protein Phosphorylation & Human Disease Group & USR3151, Station Biologique, Roscoff, France (N.L., L.M.); ManRos Therapeutics, Roscoff, France (L.M.)
| | - Laurent Meijer
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, Biomedical Research Institute (IRIB), PRES Normandy, TC2N network, University of Rouen, Mont-Saint-Aignan, France (M.J., C.L., L.D., M.-T.S., V.L., P.G., H.C.); Neuro-oncology department, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France (C.M., J.H.); Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292, Lyon, France (C.M., J.H.); University of Claude Bernard - Lyon 1, Villeurbanne, France (C.M., J.H.); Institut de Chimie et Biochimie Moléculaires et Supramoléculaires UMR 5246, University of Claude Bernard - Lyon 1, Villeurbanne, France (F.L., G.L., B.J., N.L.); Protein Phosphorylation & Human Disease Group & USR3151, Station Biologique, Roscoff, France (N.L., L.M.); ManRos Therapeutics, Roscoff, France (L.M.)
| | - Jérôme Honnorat
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, Biomedical Research Institute (IRIB), PRES Normandy, TC2N network, University of Rouen, Mont-Saint-Aignan, France (M.J., C.L., L.D., M.-T.S., V.L., P.G., H.C.); Neuro-oncology department, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France (C.M., J.H.); Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292, Lyon, France (C.M., J.H.); University of Claude Bernard - Lyon 1, Villeurbanne, France (C.M., J.H.); Institut de Chimie et Biochimie Moléculaires et Supramoléculaires UMR 5246, University of Claude Bernard - Lyon 1, Villeurbanne, France (F.L., G.L., B.J., N.L.); Protein Phosphorylation & Human Disease Group & USR3151, Station Biologique, Roscoff, France (N.L., L.M.); ManRos Therapeutics, Roscoff, France (L.M.)
| | - Pierrick Gandolfo
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, Biomedical Research Institute (IRIB), PRES Normandy, TC2N network, University of Rouen, Mont-Saint-Aignan, France (M.J., C.L., L.D., M.-T.S., V.L., P.G., H.C.); Neuro-oncology department, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France (C.M., J.H.); Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292, Lyon, France (C.M., J.H.); University of Claude Bernard - Lyon 1, Villeurbanne, France (C.M., J.H.); Institut de Chimie et Biochimie Moléculaires et Supramoléculaires UMR 5246, University of Claude Bernard - Lyon 1, Villeurbanne, France (F.L., G.L., B.J., N.L.); Protein Phosphorylation & Human Disease Group & USR3151, Station Biologique, Roscoff, France (N.L., L.M.); ManRos Therapeutics, Roscoff, France (L.M.)
| | - Hélène Castel
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, Biomedical Research Institute (IRIB), PRES Normandy, TC2N network, University of Rouen, Mont-Saint-Aignan, France (M.J., C.L., L.D., M.-T.S., V.L., P.G., H.C.); Neuro-oncology department, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France (C.M., J.H.); Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292, Lyon, France (C.M., J.H.); University of Claude Bernard - Lyon 1, Villeurbanne, France (C.M., J.H.); Institut de Chimie et Biochimie Moléculaires et Supramoléculaires UMR 5246, University of Claude Bernard - Lyon 1, Villeurbanne, France (F.L., G.L., B.J., N.L.); Protein Phosphorylation & Human Disease Group & USR3151, Station Biologique, Roscoff, France (N.L., L.M.); ManRos Therapeutics, Roscoff, France (L.M.)
| |
Collapse
|
19
|
Villicaña C, Cruz G, Zurita M. The basal transcription machinery as a target for cancer therapy. Cancer Cell Int 2014; 14:18. [PMID: 24576043 PMCID: PMC3942515 DOI: 10.1186/1475-2867-14-18] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 02/21/2014] [Indexed: 01/11/2023] Open
Abstract
General transcription is required for the growth and survival of all living cells. However, tumor cells require extraordinary levels of transcription, including the transcription of ribosomal RNA genes by RNA polymerase I (RNPI) and mRNA by RNA polymerase II (RNPII). In fact, cancer cells have mutations that directly enhance transcription and are frequently required for cancer transformation. For example, the recent discovery that MYC enhances the transcription of the majority genes in the genome correlates with the fact that several transcription interfering drugs preferentially kill cancer cells. In recent years, advances in the mechanistic studies of the basal transcription machinery and the discovery of drugs that interfere with multiple components of transcription are being used to combat cancer. For example, drugs such as triptolide that targets the general transcription factors TFIIH and JQ1 to inhibit BRD4 are administered to target the high proliferative rate of cancer cells. Given the importance of finding new strategies to preferentially sensitize tumor cells, this review primarily focuses on several transcription inhibitory drugs to demonstrate that the basal transcription machinery constitutes a potential target for the design of novel cancer drugs. We highlight the drugs’ mechanisms for interfering with tumor cell survival, their importance in cancer treatment and the challenges of clinical application.
Collapse
Affiliation(s)
| | | | - Mario Zurita
- Departament of Developmental Genetics, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Mexico, Mexico.
| |
Collapse
|
20
|
Li XQ, Ouyang ZG, Zhang SH, Liu H, Shang Y, Li Y, Zhen YS. Synergistic inhibition of angiogenesis and glioma cell-induced angiogenesis by the combination of temozolomide and enediyne antibiotic lidamycin. Cancer Biol Ther 2014; 15:398-408. [PMID: 24424202 DOI: 10.4161/cbt.27626] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Present work mainly evaluated the inhibitory effects of lidamycin (LDM), an enediyne antibiotic, on angiogenesis or glioma-induced angiogenesis in vitro and in vivo, especially its synergistic anti-angiogenesis with temozolomide (TMZ). LDM alone efficiently inhibited proliferations and induced apoptosis of rat brain microvessel endothelial cells (rBMEC). LDM also interrupted the tube formation of rat brain microvessel endothelial cells (rBMEC) and rat aortic ring spreading. The blockade of rBMEC invasion and C6 cell-induced rBMEC migration by LDM was associated with decrease of VEGF secretion in a co-culture system. TMZ dramatically potentiated the effects of LDM on anti-proliferation, apoptosis induction, and synergistically inhibited angiogenesis events. As determined by western blot and ELISA, the interaction of tumor cells and the rBMEC was markedly interrupted by LDM plus TMZ with synergistic regulations of VEGF induced angiogenesis signal pathway, tumor cell invasion/migration, and apoptosis signal pathway. Immunofluorohistochemistry of CD31 and VEGF showed that LDM plus TMZ resulted in synergistic decrease of microvessel density (MVD) and VEGF expression in human glioma U87 cell subcutaneous xenograft. This study indicates that the high efficacy of LDM and the synergistic effects of LDM plus TMZ against glioma are mediated, at least in part, by the potentiated anti-angiogenesis.
Collapse
Affiliation(s)
- Xing-Qi Li
- College of Life Science & Technology; Heilongjiang Bayi Agricultural University; Daqing, PR China; Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing, PR China
| | - Zhi-Gang Ouyang
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing, PR China
| | - Sheng-Hua Zhang
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing, PR China
| | - Hong Liu
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing, PR China
| | - Yue Shang
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing, PR China
| | - Yi Li
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing, PR China
| | - Yong-Su Zhen
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing, PR China
| |
Collapse
|
21
|
Testing of SNS-032 in a Panel of Human Neuroblastoma Cell Lines with Acquired Resistance to a Broad Range of Drugs. Transl Oncol 2013; 6:685-96. [PMID: 24466371 DOI: 10.1593/tlo.13544] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 09/29/2013] [Accepted: 09/30/2013] [Indexed: 02/07/2023] Open
Abstract
Novel treatment options are needed for the successful therapy of patients with high-risk neuroblastoma. Here, we investigated the cyclin-dependent kinase (CDK) inhibitor SNS-032 in a panel of 109 neuroblastoma cell lines consisting of 19 parental cell lines and 90 sublines with acquired resistance to 14 different anticancer drugs. Seventy-three percent of the investigated neuroblastoma cell lines and all four investigated primary tumor samples displayed concentrations that reduce cell viability by 50% in the range of the therapeutic plasma levels reported for SNS-032 (<754 nM). Sixty-two percent of the cell lines and two of the primary samples displayed concentrations that reduce cell viability by 90% in this concentration range. SNS-032 also impaired the growth of the multidrug-resistant cisplatin-adapted UKF-NB-3 subline UKF-NB-3(r)CDDP(1000) in mice. ABCB1 expression (but not ABCG2 expression) conferred resistance to SNS-032. The antineuroblastoma effects of SNS-032 did not depend on functional p53. The antineuroblastoma mechanism of SNS-032 included CDK7 and CDK9 inhibition-mediated suppression of RNA synthesis and subsequent depletion of antiapoptotic proteins with a fast turnover rate including X-linked inhibitor of apoptosis (XIAP), myeloid cell leukemia sequence 1 (Mcl-1), baculoviral IAP repeat containing 2 (BIRC2; cIAP-1), and survivin. In conclusion, CDK7 and CDK9 represent promising drug targets and SNS-032 represents a potential treatment option for neuroblastoma including therapy-refractory cases.
Collapse
|
22
|
Jung HJ, Kim Y, Chang J, Kang SW, Kim JH, Kwon HJ. Mitochondrial UQCRB regulates VEGFR2 signaling in endothelial cells. J Mol Med (Berl) 2013; 91:1117-28. [DOI: 10.1007/s00109-013-1049-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 03/24/2013] [Accepted: 04/29/2013] [Indexed: 11/25/2022]
|
23
|
The impact of CDK inhibition in human malignancies associated with pronounced defects in apoptosis: advantages of multi-targeting small molecules. Future Med Chem 2012; 4:395-424. [DOI: 10.4155/fmc.12.12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Malignant cells in chronic lymphocytic leukemia (CLL) and related diseases are heterogeneous and consist primarily of long-lived resting cells in the periphery and a minor subset of dividing cells in proliferating centers. Both cell populations have different molecular signatures that play a major role in determining their sensitivity to therapy. Contemporary approaches to treating CLL are heavily reliant on cytotoxic chemotherapeutics. However, none of the current treatment regimens can be considered curative. Pharmacological CDK inhibitors have extended the repertoire of potential drugs for CLL. Multi-targeted CDK inhibitors affect CDKs involved in regulating both cell cycle progression and transcription. Their interference with transcriptional elongation represses anti-apoptotic proteins and, thus, promotes the induction of apoptosis. Importantly, there is evidence that treatment with CDK inhibitors can overcome resistance to therapy. The pharmacological CDK inhibitors have great potential for use in combination with other therapeutics and represent promising tools for the development of new curative treatments for CLL.
Collapse
|
24
|
Cui P, Yu M, Peng X, Dong L, Yang Z. Melatonin prevents human pancreatic carcinoma cell PANC-1-induced human umbilical vein endothelial cell proliferation and migration by inhibiting vascular endothelial growth factor expression. J Pineal Res 2012; 52:236-43. [PMID: 21913973 DOI: 10.1111/j.1600-079x.2011.00933.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Melatonin is an important natural oncostatic agent, and our previous studies have found its inhibitory action on tumor angiogenesis, but the mechanism remains unclear. It is well known that vascular endothelial growth factor (VEGF) plays key roles in tumor angiogenesis and has become an important target for antitumor therapy. Pancreatic cancer is a representative of the most highly vascularized and angiogenic solid tumors, which responds poorly to chemotherapy and radiation. Thus, seeking new treatment strategies targeting which have anti-angiogenic capability is urgent in clinical practice. In this study, a co-culture system between human umbilical vein endothelial cells (HUVECs) and pancreatic carcinoma cells (PANC-1) was used to investigate the direct effect of melatonin on the tumor angiogenesis and its possible action on VEGF expression. We found HUVECs exhibited an increased cell proliferation and cell migration when co-cultured with PANC-1 cells, but the process was prevented when melatonin added to the incubation medium. Melatonin at concentrations of 1 μm and 1 mm inhibited the cell proliferation and migration of HUVECs and also decreased both the VEGF protein secreted to the cultured medium and the protein produced by the PANC-1 cells. In addition, the VEGF mRNA expression was also down-regulated by melatonin. Taken together, our present study shows that melatonin at pharmacological concentrations inhibited the elevated cell proliferation and cell migration of HUVECs stimulated by co-culturing them with PANC-1 cells; this was associated with a suppression of VEGF expression in PANC-1 cells.
Collapse
Affiliation(s)
- Peilin Cui
- Department of Gastroenterology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | | | | | | | | |
Collapse
|
25
|
Kryštof V, Rárová L, Liebl J, Zahler S, Jorda R, Voller J, Cankař P. The selective P-TEFb inhibitor CAN508 targets angiogenesis. Eur J Med Chem 2011; 46:4289-94. [PMID: 21777997 DOI: 10.1016/j.ejmech.2011.06.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 06/20/2011] [Accepted: 06/27/2011] [Indexed: 10/18/2022]
Abstract
Small molecule inhibitors of cyclin-dependent kinases (CDK) have been developed as anticancer drugs with cytostatic and cytotoxic properties, but some of them have also been shown to limit angiogenesis. Here, we report that the 3,5-diaminopyrazole CAN508 inhibits endothelial cell migration and tube formation. In addition, it reduces phosphorylation of the C-terminus of RNA polymerase II and inhibits mRNA synthesis in endothelial cells, in accordance with previous observations that it has high selectivity towards the positive transcriptional regulator P-TEFb. Moreover, CAN508 reduces expression of vascular endothelial growth factor by several human cancer cell lines. The findings suggest that P-TEFb may be an attractive target for anti-angiogenic therapy.
Collapse
Affiliation(s)
- Vladimír Kryštof
- Laboratory of Growth Regulators, Faculty of Science, Palacký University & Institute of Experimental Botany ASCR, Šlechtitelů 11, 783 71 Olomouc, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
26
|
The hallmarks of CDKN1C (p57, KIP2) in cancer. Biochim Biophys Acta Rev Cancer 2011; 1816:50-6. [PMID: 21447370 DOI: 10.1016/j.bbcan.2011.03.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 03/18/2011] [Accepted: 03/22/2011] [Indexed: 12/18/2022]
Abstract
Cyclin-dependent kinase inhibitor 1C CDKN1C (p57(KIP2)) regulates several hallmarks of cancer, including apoptosis, cell invasion and metastasis, tumor differentiation and angiogenesis. p57(KIP2) is generally not mutated in cancer, but its expression is downregulated through epigenetic changes such as DNA methylation and repressive histone marks at the promoter. This opens up possibilities for therapeutic intervention through reactivation of p57(KIP2) gene expression. Furthermore, p57(KIP2) has been tested as a prognostic factor for many types of cancer, even differentiating between early and late stage cancer. In this review, the multifunctional tumor suppressor capabilities of p57(KIP2), the mechanisms of p57(KIP2) transcriptional repression in cancer, and the therapeutic potential of reactivation of p57(KIP2) protein expression will be discussed.
Collapse
|
27
|
Abstract
The pathophysiology of multiple myeloma-induced angiogenesis is complex and involves both direct production of angiogenic cytokines by plasma cells and their induction within the microenvironment. In this research, we investigated whether mesenchymal stem cells participated in inducing the angiogenic response in multiple myeloma, and explored the mechanism by which MSCs influence myeloma angiogenesis. We detected the concentration of angiogenic factors (bFGF, HGF, and VEGF) in the conditioned medium of mesenchymal stem cells and the capillary formation ability of mesenchymal stem cells in vitro. We found that conditioned medium of MSCs derived from MM significantly promoted the proliferation, chemotaxis, and capillary formation of human umbilical vein endothelial cells compared with that from normal donors. ELISA and RT-PCR were used to detect the mRNA and protein levels of angiogenic factors (bFGF, HGF, and VEGF) in the conditioned medium. We found that mRNA and protein levels of angiogenic factors were elevated in MSCs from multiple myeloma compared with normal donors.
Collapse
Affiliation(s)
- Xiaofang Wang
- Department of Hematology, Cancer Hospital of Tianjin, Tianjin Medical University, Tianjin, China.
| | | | | |
Collapse
|
28
|
Walsby E, Lazenby M, Pepper C, Burnett AK. The cyclin-dependent kinase inhibitor SNS-032 has single agent activity in AML cells and is highly synergistic with cytarabine. Leukemia 2011; 25:411-9. [PMID: 21212792 DOI: 10.1038/leu.2010.290] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
SNS-032 (BMS-387032) is a selective cyclin-dependent kinase (CDK) inhibitor. In this study, we evaluated its effects on primary acute myeloid leukemia (AML) samples (n=87). In vitro exposure to SNS-032 for 48 h resulted in a mean LD(50) of 139±203 nM; Cytarabine (Ara-C) was more than 35 times less potent in the same cohort. SNS-032-induced a dose-dependent increase in annexin V staining and caspase-3 activation. At the molecular level, SNS-032 induced a marked dephosphorylation of serine 2 and 5 of RNA polymerase (RNA Pol) II and inhibited the expression of CDK2 and CDK9 and dephosphorylated CDK7. Furthermore, the combination of SNS-032 and Ara-C showed remarkable synergy that was associated with reduced mRNA levels of the antiapoptotic genes XIAP, BCL2 and MCL1. In conclusion, SNS-032 is effective as a single agent and in combination with Ara-C in primary AML blasts. Treatment with Ara-C alone significantly induced the transcription of the antiapoptotic genes BCL2 and XIAP. In contrast, the combination of SNS-032 and Ara-C suppressed the transcription of BCL2, XIAP and MCL1. Therefore, the combination of SNS-032 and Ara-C may increase the sensitivity of AML cells to the cytotoxic effects of Ara-C by inhibiting the transcription of antiapoptotic genes.
Collapse
Affiliation(s)
- E Walsby
- Cardiff Experimental Cancer Medicine Centre, Department of Haematology, School of Medicine, Cardiff University, Heath Park, Cardiff, UK.
| | | | | | | |
Collapse
|
29
|
Sun ZJ, Chen G, Zhang W, Hu X, Huang CF, Wang YF, Jia J, Zhao YF. Mammalian Target of Rapamycin Pathway Promotes Tumor-Induced Angiogenesis in Adenoid Cystic Carcinoma: Its Suppression by Isoliquiritigenin through Dual Activation of c-Jun NH2-Terminal Kinase and Inhibition of Extracellular Signal-Regulated Kinase. J Pharmacol Exp Ther 2010; 334:500-12. [DOI: 10.1124/jpet.110.167692] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
30
|
Tong WG, Chen R, Plunkett W, Siegel D, Sinha R, Harvey RD, Badros AZ, Popplewell L, Coutre S, Fox JA, Mahadocon K, Chen T, Kegley P, Hoch U, Wierda WG. Phase I and pharmacologic study of SNS-032, a potent and selective Cdk2, 7, and 9 inhibitor, in patients with advanced chronic lymphocytic leukemia and multiple myeloma. J Clin Oncol 2010; 28:3015-22. [PMID: 20479412 DOI: 10.1200/jco.2009.26.1347] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE SNS-032 is a highly selective and potent inhibitor of cyclin-dependent kinases (Cdks) 2, 7, and 9, with in vitro growth inhibitory effects and ability to induce apoptosis in malignant B cells. A phase I dose-escalation study of SNS-032 was conducted to evaluate safety, pharmacokinetics, biomarkers of mechanism-based pharmacodynamic (PD) activity, and clinical efficacy. PATIENTS AND METHODS Parallel cohorts of previously treated patients with chronic lymphocytic leukemia (CLL) and multiple myeloma (MM) received SNS-032 as a loading dose followed by 6-hour infusion weekly for 3 weeks of each 4-week course. RESULTS There were 19 patients with CLL and 18 with MM treated. Tumor lysis syndrome was the dose-limiting toxicity (DLT) for CLL, the maximum-tolerated dose (MTD) was 75 mg/m(2), and the most frequent grade 3 to 4 toxicity was myelosuppression. One patient with CLL had more than 50% reduction in measurable disease without improvement in hematologic parameters. Another patient with low tumor burden had stable disease for four courses. For patients with MM, no DLT was observed and MTD was not identified at up to 75 mg/m(2), owing to early study closure. Two patients with MM had stable disease and one had normalization of spleen size with treatment. Biomarker analyses demonstrated mechanism-based PD activity with inhibition of Cdk7 and Cdk9, decreases in Mcl-1 and XIAP expression level, and associated CLL cell apoptosis. CONCLUSION SNS-032 demonstrated mechanism-based target modulation and limited clinical activity in heavily pretreated patients with CLL and MM. Further single-agent, PD-based, dose and schedule modification is warranted to maximize clinical efficacy.
Collapse
Affiliation(s)
- Wei-Gang Tong
- The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
The War on Cancer rages on. Neoplasia 2010; 11:1252-63. [PMID: 20019833 DOI: 10.1593/neo.91866] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 11/03/2009] [Accepted: 11/03/2009] [Indexed: 02/08/2023] Open
Abstract
In 1971, the "War on Cancer" was launched by the US government to cure cancer by the 200-year anniversary of the founding of the United States of America, 1976. This article briefly looks back at the progress that has been made in cancer research and compares progress made in other areas of human affliction. While progress has indeed been made, the battle continues to rage on.
Collapse
|
32
|
Christian BA, Grever MR, Byrd JC, Lin TS. Flavopiridol in chronic lymphocytic leukemia: a concise review. ACTA ACUST UNITED AC 2010; 9 Suppl 3:S179-85. [PMID: 19778838 DOI: 10.3816/clm.2009.s.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Patients with chronic lymphocytic leukemia (CLL) with high-risk cytogenetic features such as del(17p13) have limited treatment options and decreased overall survival. Dysfunction of p53 leads to resistance to fludarabine-based therapies. Cyclin-dependent kinase inhibitors (CDKi) are a novel class of agents that induce apoptosis in CLL cells independent of p53 mutational status. The synthetic flavone flavopiridol demonstrated promising in vitro activity in CLL. In initial phase I studies using a continuous infusion dosing schedule in a variety of malignancies, no clinical activity was observed. Detailed pharmacokinetic modeling led to the development of a novel dosing schedule designed to achieve target drug concentrations in vivo. In phase I testing, this dosing schedule resulted in acute tumor lysis syndrome (TLS) as the dose-limiting toxicity. With the implementation of a standardized protocol to prevent severe TLS, flavopiridol was administered safely, and responses were observed in heavily pretreated, fludarabine-refractory patients, cytogenetically high-risk patients, and patients with bulky lymphadenopathy. In a pharmacokinetic analysis, flavopiridol area under the plasma concentration-time curve (AUC) correlated with clinical response and cytokine release syndrome. Phase II studies are under way with encouraging preliminary results. Flavopiridol is currently under active investigation in combination with other agents and as a means to eradicate minimal residual disease in patients following cytoreductive chemotherapy. Several other investigational CDKi in preclinical and early clinical development are briefly discussed in this review.
Collapse
Affiliation(s)
- Beth A Christian
- Division of Hematology-Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA.
| | | | | | | |
Collapse
|
33
|
Abstract
Progression of the cell cycle is controlled by various activating and inhibiting cellular factors. The subtle balance between these counteracting regulators in normal cells ensures proper cell cycle progression and facilitates cellular responses to a variety of stress stimuli. Key activators include cyclin-dependent kinases (CDKs) and, consequently, loss or inactivation of CDK inhibitors contributes to the escape of cancer cells from cell cycle control and hyperactivation of CDKs occurs in various neurodegenerative disorders. However, these adverse effects may be compensated by pharmacological counterparts. Inhibitors of CDKs representing various classes of compounds with diverse CDK inhibitory patterns have been developed, but inhibitors that have high selectivity and offer highly targeted activity against both cell cycle and transcriptional CDKs are of particular interest. This review focuses on pharmacological CDK inhibitors that have entered clinical trials and some compounds that have been evaluated preclinically. Recent discoveries in cell cycle regulation have provided rationales for clinical applications of CDK inhibitors in both monotherapeutic and combined therapeutic regimens.
Collapse
|
34
|
Boquoi A, Chen T, Enders GH. Chemoprevention of mouse intestinal tumorigenesis by the cyclin-dependent kinase inhibitor SNS-032. Cancer Prev Res (Phila) 2009; 2:800-6. [PMID: 19723896 DOI: 10.1158/1940-6207.capr-09-0053] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Despite advances in screening and treatment, colorectal cancer remains the second leading cause of cancer-related death in the United States. Cyclin-dependent kinases (Cdk) are deregulated in colorectal cancer by silencing of the Cdk inhibitor p16(Ink4a) and other mechanisms. We tested whether the small molecule Cdk inhibitor SNS-032 (formerly BMS-387032), which targets Cdk2, Cdk7, and Cdk9, can prevent intestinal tumorigenesis in mouse models. We generated mice with high intestinal tumor loads by combining the multiple intestinal neoplasia (Min) mutation with Ink4a/Arf mutations and inducing colitis with dextran sulfate sodium. p16-null Min mice (n = 17) began dextran sulfate sodium treatment at week 5 and i.p. injection of carrier or SNS-032 at week 6. Mice were sacrificed at week 12. SNS-032 was well tolerated and reduced colon tumor burden to 36% of that in carrier-treated mice (P < 0.001). We then extended the study to Ink4/Arf-null Min mice (n = 14) and increased the drug dose frequency. SNS-032 treatment reduced the intestinal tumor number to 25% and intestinal tumor burden to 16% of carrier-treated mice (P < 0.0001). DNA synthesis in non-neoplastic and tumor epithelial cells, detected by bromodeoxyuridine incorporation, was modestly reduced by acute SNS-032 treatment. The mitotic index, detected by histone H3 phosphorylation, was distinctly decreased (P < 0.03), and apoptosis, detected by caspase 3 activation, was increased (P < 0.005). These results show the chemoprevention of intestinal tumorigenesis by SNS-032. Our findings support further study of Cdk inhibitors for chemoprevention and therapy of colon cancer.
Collapse
Affiliation(s)
- Amelie Boquoi
- Department of Medicine, Epigenetics and Progenitor Cell Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | |
Collapse
|
35
|
The hypoxia-inducible factor 1/NOR-1 axis regulates the survival response of endothelial cells to hypoxia. Mol Cell Biol 2009; 29:5828-42. [PMID: 19720740 DOI: 10.1128/mcb.00945-09] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hypoxia induces apoptosis but also triggers adaptive mechanisms to ensure cell survival. Here we show that the prosurvival effects of hypoxia-inducible factor 1 (HIF-1) in endothelial cells are mediated by neuron-derived orphan receptor 1 (NOR-1). The overexpression of NOR-1 decreased the rate of endothelial cells undergoing apoptosis in cultures exposed to hypoxia, while the inhibition of NOR-1 increased cell apoptosis. Hypoxia upregulated NOR-1 mRNA levels in a time- and dose-dependent manner. Blocking antibodies against VEGF or SU5614 (a VEGF receptor 2 inhibitor) did not prevent hypoxia-induced NOR-1 expression, suggesting that NOR-1 is not induced by the autocrine secretion of VEGF in response to hypoxia. The reduction of HIF-1 alpha protein levels by small interfering RNAs, or by inhibitors of the phosphatidylinositol-3 kinase (PI3K)/Akt pathway or mTOR, significantly counteracted hypoxia-induced NOR-1 upregulation. Intracellular Ca(2+) was involved in hypoxia-induced PI3K/Akt activation and in the downstream NOR-1 upregulation. A hypoxia response element mediated the transcriptional activation of NOR-1 induced by hypoxia as we show by transient transfection and chromatin immunoprecipitation assays. Finally, the attenuation of NOR-1 expression reduced both basal and hypoxia-induced cIAP2 (cellular inhibitor of apoptosis protein 2) mRNA levels, while NOR-1 overexpression upregulated cIAP2. Therefore, NOR-1 is a downstream effector of HIF-1 signaling involved in the survival response of endothelial cells to hypoxia.
Collapse
|
36
|
Neoplasia: the second decade. Neoplasia 2009; 10:1314-24. [PMID: 19048110 DOI: 10.1593/neo.81372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Revised: 10/27/2008] [Accepted: 10/27/2008] [Indexed: 12/30/2022] Open
Abstract
This issue marks the end of the 10-year anniversary of Neoplasia where we have seen exciting growth in both number of submitted and published articles in Neoplasia. Neoplasia was first published in 1999. During the past 10 years, Neoplasia has dynamically adapted to the needs of the cancer research community as technologies have advanced. Neoplasia is currently providing access to articles through PubMed Central to continue to facilitate rapid broad-based dissemination of published findings to the scientific community through an Open Access model. This has in part helped Neoplasia to achieve an improved impact factor this past year, demonstrating that the manuscripts published by Neoplasia are of great interest to the overall cancer research community. This past year, Neoplasia received a record number of articles for review and has had a 21% increase in the number of published articles.
Collapse
|
37
|
Combining bevacizumab with temozolomide increases the antitumor efficacy of temozolomide in a human glioblastoma orthotopic xenograft model. Neoplasia 2009; 10:1383-92. [PMID: 19048117 DOI: 10.1593/neo.08928] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Revised: 09/09/2008] [Accepted: 09/12/2008] [Indexed: 01/20/2023] Open
Abstract
PURPOSE The aims of the present work were to investigate the in vitro and in vivo antiangiogenic effects of chronic temozolomide treatment on various glioma models and to demonstrate whether bevacizumab (Avastin) increased the therapeutic benefits contributed by temozolomide in glioma. EXPERIMENTAL DESIGN The expression levels of various antiangiogenic factors in four glioma cell lines were evaluated after chronic in vitro treatment with temozolomide by Western blot. Proliferation and migration assays were performed on human endothelial cells incubated with supernatants of glioma cells treated with and without temozolomide. Orthotopic glioma models were used to evaluate the antiangiogenic effects of temozolomide in vivo and the therapeutic benefits of different temozolomide treatment schedules used alone or in combination with bevacizumab. RESULTS Temozolomide, a proautophagic and proapoptotic drug, decreased the expression levels of HIF-1alpha, ID-1, ID-2, and cMyc in the glioma models investigated, all of which playing major roles in angiogenesis and the switch to hypoxic metabolism. These changes could be, at least partly, responsible for the impairment of angiogenesis observed in vitro and in vivo. Moreover, combining bevacizumab with temozolomide increased the survival of glioma-bearing mice in comparison to each compound administered alone. CONCLUSIONS In addition to the numerous mechanisms of action already identified for temozolomide, we report here that it also exerts antitumor effects by impairing angiogenic processes. We further emphasize that bevacizumab, which is an antiangiogenic drug with a different mechanism of action, could be useful in combination with temozolomide to increase the latter's therapeutic benefit in glioma patients.
Collapse
|
38
|
Kodym E, Kodym R, Reis AE, Habib AA, Story MD, Saha D. The small-molecule CDK inhibitor, SNS-032, enhances cellular radiosensitivity in quiescent and hypoxic non-small cell lung cancer cells. Lung Cancer 2009; 66:37-47. [PMID: 19193471 DOI: 10.1016/j.lungcan.2008.12.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Revised: 12/18/2008] [Accepted: 12/22/2008] [Indexed: 10/21/2022]
Abstract
In solid tumors, including non-small cell lung carcinomas (NSCLC) the existence of radioresistant subpopulations, such as quiescent or hypoxic tumor cells, is well established, thus posing a critical therapeutic problem. Although small-molecule inhibitors targeting cyclin-dependent kinases (CDK) were demonstrated to enhance cellular radiosensitivity preferentially in proliferating tumor cells, cell cycle-independent activities of these substances were recently suggested. In this study, the potential of a newer generation small-molecule CDK inhibitor, SNS-032, to sensitize radioresistant tumor cells to ionizing radiation was tested in vitro using two NSCLC cell lines (NCI-H460 and A549). Exposure of quiescent and hypoxic lung tumor cells to SNS-032 at a clinically achievable concentration (500 nM) prior to irradiation resulted in a significant increase in cellular radiosensitivity indicating cell cycle-unrelated mechanisms. The effect of SNS-032 on non-cycling cells was not attributed to an enhanced toxicity of the drug. A SNS-032 mediated delay in the resolution of radiation-induced gammaH2AX foci a surrogate for DNA double-strand breaks was determined in non-cycling cells, suggesting a modulation of DNA double-strand break repair. These results indicate a modulation of DNA double-strand break repair to be partially attributed to the radiosensitization effects of SNS-032 observed in hypoxic and quiescent lung tumor cells. Considering the importance of therapy resistance for the radiocurability of solid tumors, our findings may provide the basis for an improvement of the well-established treatment regimens in clinical oncology.
Collapse
Affiliation(s)
- Elisabeth Kodym
- Department of Radiation Oncology, Division of Molecular Radiation Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9187, USA
| | | | | | | | | | | |
Collapse
|
39
|
Conroy A, Stockett DE, Walker D, Arkin MR, Hoch U, Fox JA, Hawtin RE. SNS-032 is a potent and selective CDK 2, 7 and 9 inhibitor that drives target modulation in patient samples. Cancer Chemother Pharmacol 2009; 64:723-32. [PMID: 19169685 DOI: 10.1007/s00280-008-0921-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Accepted: 12/26/2008] [Indexed: 10/21/2022]
Abstract
PURPOSE SNS-032 (formerly BMS-387032) is a potent, selective inhibitor of cyclin-dependent kinases (CDK) 2, 7 and 9, currently in phase 1 clinical trial for chronic lymphocytic leukemia (CLL) and multiple myeloma (MM). We used the MM cell line RPMI-8226 to evaluate the relationship between duration of SNS-032 exposure, target modulation of CDKs 2, 7 and 9, and induction of apoptosis. We also assessed target modulation in patient peripheral blood mononuclear cells (PBMCs) from phase 1 solid tumor patients treated with SNS-032. METHODS Proliferation and colony forming assays were used to evaluate cytotoxicity, Western blot analyses to evaluate target modulation, FACS analysis to assess cell cycle distribution, RT-PCR to evaluate transcriptional inhibition. RESULTS SNS-032 blocks the cell cycle via inhibition of CDKs 2 and 7, and transcription via inhibition of CDKs 7 and 9. Treatment of RPMI-8226 MM cells at 300 nM (IC(90)) for 6 h was sufficient for commitment to apoptosis. This correlated with inhibition of CDKs 2, 7 and 9, as reflected in substrate signaling molecules. SNS-032 activity was unaffected by human serum. Target modulation was observed in PBMC from treated patients. CONCLUSIONS These results demonstrate SNS-032 target modulation of CDKs 2, 7 and 9, and establish 6 h exposure as sufficient to commit RPMI-8226 MM cells to apoptosis. Combined with the demonstration of target modulation in PBMC from phase 1 solid tumor patients treated with SNS-032, these data support the ongoing clinical study of SNS-032 in MM and CLL.
Collapse
Affiliation(s)
- Andrew Conroy
- Sunesis Pharmaceuticals Inc, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Neoplasia: An Anniversary of Progress. Neoplasia 2007. [DOI: 10.1593/neo.07968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|