1
|
Yevale D, Teraiya N, Lalwani T, Dalasaniya M, Patel SK, Dixit N, Sangani CB, Kumar S, Mulukuri NVLS, Huang T, Duan YT, Zhang J. Discovery of new pyrazole-4-carboxamide analogues as potential anticancer agents targeting dual aurora kinase A and B. Eur J Med Chem 2024; 280:116917. [PMID: 39388904 DOI: 10.1016/j.ejmech.2024.116917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024]
Abstract
Aurora kinases A and B are critical regulators of cell division and cytokinesis. Abnormal expression of Aurora kinases A and B causes chromosomal instability and disrupts several tumor suppressor and oncoprotein-controlled pathways. As a result, there has been a spike in interest in developing inhibitors against these kinases as anticancer treatments. This paper addresses the discovery, anticancer evaluation and druggability study of new pyrazole-4-carboxamide analogues as kinases inhibitors. Among the compounds, 6k demonstrated the highest cytotoxicity against HeLa and HepG2 cells, with IC50 of 0.43 μM and 0.67 μM, respectively. It selectively inhibited Aurora kinases A and B, with IC50 values of 16.3 nM and 20.2 nM, respectively, in comparison to other kinases. Molecular investigations revealed that 6k induced the inhibition of phosphorylated Thr288 (Aurora kinase A) and phosphorylated Histone H3 (Aurora kinase B), confirming its mechanism of action. Beside, compound 6k arrested the cell cycle at the G2/M phase by modulating cyclinB1 and cdc2 protein levels and increasing the Sub-G1 cell population. It also significantly increased polyploidization (>8 N) and abnormal mitosis, likely due to Aurora kinase inhibition. Furthermore, 6k boosted apoptosis through the intrinsic route, with elevated levels of p53, Bak, Bax, cleaved caspase-3, and cleaved PARP. Moreover, docking and MD simulations validated kinase inhibition-induced anticancer effects. Additionally, 6k satisfied drug-likeness parameters and remained stable in the in vitro metabolism. These findings indicate that 6k warrants further in vivo pharmacokinetic and pharmacodynamics investigations.
Collapse
Affiliation(s)
- Digambar Yevale
- Henan Provincial Key Laboratory of Pediatric Hematology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China; Department of Chemistry, Shri M.M Patel Institute of Sciences and Research, Kadi Sarva Vishwavidyalaya, Gandhinagar, Gujarat, 382016, India
| | - Nishith Teraiya
- Department of Pharmaceutical Chemistry, K B Institute of Pharmaceutical Education and Research, Kadi Sarva Vishvavidhyalay, Gandhinagar, Gujarat, 382023, India
| | - Twinkle Lalwani
- Piramal Pharma Limited, Plot No. 18, Pharmaceutical Special Economic Zone, Village-Matoda, Taluka- Sanand, Ahmedabad, Gujarat, 382213, India
| | - Mayur Dalasaniya
- Piramal Pharma Limited, Plot No. 18, Pharmaceutical Special Economic Zone, Village-Matoda, Taluka- Sanand, Ahmedabad, Gujarat, 382213, India
| | - Saumya K Patel
- Department of Botany, Bioinformatics and Climate Change Impacts Management, School of Science, Gujarat University, Ahmedabad, Gujarat, 380009, India
| | - Nandan Dixit
- Department of Botany, Bioinformatics and Climate Change Impacts Management, School of Science, Gujarat University, Ahmedabad, Gujarat, 380009, India
| | - Chetan B Sangani
- Department of Chemistry, Government Science College, Sector-15, Gandhinagar, Gujarat University, Gujarat, 382016, India.
| | - Sujeet Kumar
- Department of Pharmaceutical Chemistry and Pharmacognosy, Nitte College of Pharmaceutical Sciences, Bangalore, Karnataka, 560064, India
| | - N V L Sirisha Mulukuri
- Department of Pharmaceutical Chemistry and Pharmacognosy, Nitte College of Pharmaceutical Sciences, Bangalore, Karnataka, 560064, India
| | - Tao Huang
- Medical School, Huanghe Science and Technology University, Zhengzhou, China
| | - Yong-Tao Duan
- Henan Provincial Key Laboratory of Pediatric Hematology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China.
| | - Jie Zhang
- Henan Provincial Key Laboratory of Pediatric Hematology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China.
| |
Collapse
|
2
|
Zemlianski V, Marešová A, Princová J, Holič R, Häsler R, Ramos Del Río MJ, Lhoste L, Zarechyntsava M, Převorovský M. Nitrogen availability is important for preventing catastrophic mitosis in fission yeast. J Cell Sci 2024; 137:jcs262196. [PMID: 38780300 DOI: 10.1242/jcs.262196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Abstract
Mitosis is a crucial stage in the cell cycle, controlled by a vast network of regulators responding to multiple internal and external factors. The fission yeast Schizosaccharomyces pombe demonstrates catastrophic mitotic phenotypes due to mutations or drug treatments. One of the factors provoking catastrophic mitosis is a disturbed lipid metabolism, resulting from, for example, mutations in the acetyl-CoA/biotin carboxylase (cut6), fatty acid synthase (fas2, also known as lsd1) or transcriptional regulator of lipid metabolism (cbf11) genes, as well as treatment with inhibitors of fatty acid synthesis. It has been previously shown that mitotic fidelity in lipid metabolism mutants can be partially rescued by ammonium chloride supplementation. In this study, we demonstrate that mitotic fidelity can be improved by multiple nitrogen sources. Moreover, this improvement is not limited to lipid metabolism disturbances but also applies to a number of unrelated mitotic mutants. Interestingly, the partial rescue is not achieved by restoring the lipid metabolism state, but rather indirectly. Our results highlight a novel role for nitrogen availability in mitotic fidelity.
Collapse
Affiliation(s)
- Viacheslav Zemlianski
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00 Prague 2, Czechia
| | - Anna Marešová
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00 Prague 2, Czechia
| | - Jarmila Princová
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00 Prague 2, Czechia
| | - Roman Holič
- Centre of Biosciences SAS, Institute of Animal Biochemistry and Genetics, Dúbravská cesta 9, 840 05 Bratislava, Slovak Republic
| | - Robert Häsler
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Rosalind-Franklin-Straße 9, 24105 Kiel, Germany
| | - Manuel José Ramos Del Río
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00 Prague 2, Czechia
| | - Laurane Lhoste
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00 Prague 2, Czechia
| | - Maryia Zarechyntsava
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00 Prague 2, Czechia
| | - Martin Převorovský
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00 Prague 2, Czechia
| |
Collapse
|
3
|
Zhang W, Dong J, Wu Y, Liang X, Suo L, Wang L. Integrated Bioinformatic Analysis Reveals the Oncogenic, Survival, and Prognostic Characteristics of TPX2 in Hepatocellular Carcinoma. Biochem Genet 2024:10.1007/s10528-024-10840-3. [PMID: 38833082 DOI: 10.1007/s10528-024-10840-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 05/13/2024] [Indexed: 06/06/2024]
Abstract
Targeting protein for Xenopus kinesin-like protein 2 (TPX2), a well-known mitotic protein, has been linked to carcinogenesis in several cancers. This study investigated the role of TPX2 in hepatocellular carcinoma (HCC) from various aspects using bioinformatic analyses. TPX2 expression and its prognostic value in pan-cancers were analyzed using SangerBox. TPX2 expression and its association with prognosis, immune infiltration, tumor mutations, and signaling pathways in HCC were analyzed using UALCAN, BoxKaplan-Meier Plotter, GEPIA, Human Protein Atlas, TIMER 2.0, and SangerBox. Genes co-expressed with TPX2 in HCC were analyzed using the HCCDB database, followed by functional enrichment using SangerBox. Clinical predictive models were established based on TPX2 and its co-expressed genes using the ACLBI database. TPX2 expression significantly increased in pan-cancers and was associated with survival in nearly half of the cancer types. High TPX2 expression has been linked to poor survival outcomes in patients with HCC. TPX2 expression was positively correlated with abundant infiltration of immune cells (including B cells, CD4 + /CD8 + T cells, macrophages, neutrophils, and dendritic cells), TP53 mutation, and carcinogenesis-related pathways, such as the PI3K/AKT/mTOR pathway, cellular response to hypoxia, and tumor proliferation signature. Nineteen genes were found to be co-expressed with TPX2 in HCC, and these genes showed close positive correlations and were mainly implicated in cell cycle-related functions. A prognostic model established using TPX2 and its expressed genes could stratify HCC patients into high- and low-risk groups, with a significantly shorter survival time in high-risk groups. The prognostic model performed well in predicting 1-, 3-, and 5-year survival of patients with HCC, with areas under the curve of 0.801, 0.725, and 0.711, respectively. TPX2 functions as an oncogene in HCC, and its high expression is detrimental to the survival of patients with HCC. Thus, TPX2 may be a prognostic biomarker and potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Weibin Zhang
- Department of Hepatobiliary Surgery, The Second Hospital of Dalian Medical University, Dalian, China
| | - Jia Dong
- Department of Radiology, Jinzhou Maternity and Infant Hospital, Jinzhou, China
| | - Yunfei Wu
- Department of General Surgery, Jinzhou Central Hospital, Jinzhou, China
| | - Xiangnan Liang
- Department of Hepatobiliary Surgery, The Second Hospital of Dalian Medical University, Dalian, China
| | - Lida Suo
- Department of Hepatobiliary Surgery, The Second Hospital of Dalian Medical University, Dalian, China
| | - Liming Wang
- Department of Hepatobiliary Surgery, The Second Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
4
|
Onyiba CI, Scarlett CJ, Weidenhofer J. The Mechanistic Roles of Sirtuins in Breast and Prostate Cancer. Cancers (Basel) 2022; 14:cancers14205118. [PMID: 36291902 PMCID: PMC9600935 DOI: 10.3390/cancers14205118] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/06/2022] [Accepted: 10/14/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary There are diverse reports of the dual role of sirtuin genes and proteins in breast and prostate cancers. This review discusses the current information on the tumor promotion or suppression roles of SIRT1–7 in breast and prostate cancers. Precisely, we highlight that sirtuins regulate various proteins implicated in proliferation, apoptosis, autophagy, chemoresistance, invasion, migration, and metastasis of both breast and prostate cancer. We also provide evidence of the direct regulation of sirtuins by miRNAs, highlighting the consequences of this regulation in breast and prostate cancer. Overall, this review reveals the potential value of sirtuins as biomarkers and/or targets for improved treatment of breast and prostate cancers. Abstract Mammalian sirtuins (SIRT1–7) are involved in a myriad of cellular processes, including apoptosis, proliferation, differentiation, epithelial-mesenchymal transition, aging, DNA repair, senescence, viability, survival, and stress response. In this review, we discuss the current information on the mechanistic roles of SIRT1–7 and their downstream effects (tumor promotion or suppression) in cancers of the breast and prostate. Specifically, we highlight the involvement of sirtuins in the regulation of various proteins implicated in proliferation, apoptosis, autophagy, chemoresistance, invasion, migration, and metastasis of breast and prostate cancer. Additionally, we highlight the available information regarding SIRT1–7 regulation by miRNAs, laying much emphasis on the consequences in the progression of breast and prostate cancer.
Collapse
Affiliation(s)
- Cosmos Ifeanyi Onyiba
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Ourimbah, NSW 2258, Australia
- Correspondence:
| | - Christopher J. Scarlett
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Ourimbah, NSW 2258, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Judith Weidenhofer
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Ourimbah, NSW 2258, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
5
|
Liu K, Zheng M, Lu R, Du J, Zhao Q, Li Z, Li Y, Zhang S. The role of CDC25C in cell cycle regulation and clinical cancer therapy: a systematic review. Cancer Cell Int 2020; 20:213. [PMID: 32518522 PMCID: PMC7268735 DOI: 10.1186/s12935-020-01304-w] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 05/28/2020] [Indexed: 12/24/2022] Open
Abstract
One of the most prominent features of tumor cells is uncontrolled cell proliferation caused by an abnormal cell cycle, and the abnormal expression of cell cycle-related proteins gives tumor cells their invasive, metastatic, drug-resistance, and anti-apoptotic abilities. Recently, an increasing number of cell cycle-associated proteins have become the candidate biomarkers for early diagnosis of malignant tumors and potential targets for cancer therapies. As an important cell cycle regulatory protein, Cell Division Cycle 25C (CDC25C) participates in regulating G2/M progression and in mediating DNA damage repair. CDC25C is a cyclin of the specific phosphatase family that activates the cyclin B1/CDK1 complex in cells for entering mitosis and regulates G2/M progression and plays an important role in checkpoint protein regulation in case of DNA damage, which can ensure accurate DNA information transmission to the daughter cells. The regulation of CDC25C in the cell cycle is affected by multiple signaling pathways, such as cyclin B1/CDK1, PLK1/Aurora A, ATR/CHK1, ATM/CHK2, CHK2/ERK, Wee1/Myt1, p53/Pin1, and ASK1/JNK-/38. Recently, it has evident that changes in the expression of CDC25C are closely related to tumorigenesis and tumor development and can be used as a potential target for cancer treatment. This review summarizes the role of CDC25C phosphatase in regulating cell cycle. Based on the role of CDC25 family proteins in the development of tumors, it will become a hot target for a new generation of cancer treatments.
Collapse
Affiliation(s)
- Kai Liu
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People's Republic of China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People's Republic of China
| | - Rui Lu
- Department of Pathology, Tianjin Nankai Hospital, Tianjin, People's Republic of China
| | - Jiaxing Du
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People's Republic of China
| | - Qi Zhao
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People's Republic of China
| | - Zugui Li
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People's Republic of China
| | - Yuwei Li
- Departments of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121 People's Republic of China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People's Republic of China
| |
Collapse
|
6
|
Synthesis, biological evaluation and molecular modeling study of 2-amino-3,5-disubstituted-pyrazines as Aurora kinases inhibitors. Bioorg Med Chem 2020; 28:115351. [DOI: 10.1016/j.bmc.2020.115351] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
7
|
Weidle UH, Schmid D, Birzele F, Brinkmann U. MicroRNAs Involved in Metastasis of Hepatocellular Carcinoma: Target Candidates, Functionality and Efficacy in Animal Models and Prognostic Relevance. Cancer Genomics Proteomics 2020; 17:1-21. [PMID: 31882547 PMCID: PMC6937123 DOI: 10.21873/cgp.20163] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is responsible for the second-leading cancer-related death toll worldwide. Although sorafenib and levantinib as frontline therapy and regorafenib, cabazantinib and ramicurimab have now been approved for second-line therapy, the therapeutic benefit is in the range of only a few months with respect to prolongation of survival. Aggressiveness of HCC is mediated by metastasis. Intrahepatic metastases and distant metastasis to the lungs, lymph nodes, bones, omentum, adrenal gland and brain have been observed. Therefore, the identification of metastasis-related new targets and treatment modalities is of paramount importance. In this review, we focus on metastasis-related microRNAs (miRs) as therapeutic targets for HCC. We describe miRs which mediate or repress HCC metastasis in mouse xenograft models. We discuss 18 metastasis-promoting miRs and 35 metastasis-inhibiting miRs according to the criteria as outlined. Six of the metastasis-promoting miRs (miR-29a, -219-5p, -331-3p, 425-5p, -487a and -1247-3p) are associated with unfavourable clinical prognosis. Another set of six down-regulated miRs (miR-101, -129-3p, -137, -149, -503, and -630) correlate with a worse clinical prognosis. We discuss the corresponding metastasis-related targets as well as their potential as therapeutic modalities for treatment of HCC-related metastasis. A subset of up-regulated miRs -29a, -219-5p and -425-5p and down-regulated miRs -129-3p and -630 were evaluated in orthotopic metastasis-related models which are suitable to mimic HCC-related metastasis. Those miRNAs may represent prioritized targets emerging from our survey.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| | - Daniela Schmid
- Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| | - Fabian Birzele
- Pharmaceutical Sciences, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, Basel, Switzerland
| | - Ulrich Brinkmann
- Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
8
|
Qi B, Xu X, Yang Y, He H, Yue X. Optimization and biological evaluation of nicotinamide derivatives as Aurora kinase inhibitors. Bioorg Med Chem 2019; 27:3825-3835. [PMID: 31307762 DOI: 10.1016/j.bmc.2019.07.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 10/26/2022]
Abstract
Aurora kinases are known to be overexpressed in various solid tumors and implicated in oncogenesis and tumor progression. A series of nicotinamide derivatives were synthesized and their biological activities were evaluated, including kinase inhibitory activity against Aur A and Aur B and in vitro antitumor activity against SW620, HT-29, NCI-H1975 and Hela cancer cell lines. In addition, the study of antiproliferation, cytotoxicity and apoptosis was performed meanwhile. As the most potent inhibitor of Aur A, 4-((3-bromo-4-fluorophenyl)amino)-6-chloro-N-(4-((6,7-dimethoxyquinolin-4-yl)oxy)-3-fluorophenyl)nicotinamide (10l) showed excellent antitumor activity against SW620 and NCI-H1975 with IC50 values were 0.61 and 1.06 μM, while the IC50 values of reference compound were 3.37 and 6.67 μM, respectively. Furthermore, binding mode studies indicated that compound 10l forms better interaction with Aur A.
Collapse
Affiliation(s)
- Baohui Qi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, Guangdong Province, China.
| | - Xingwei Xu
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, Guangdong Province, China
| | - Ying Yang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, Guangdong Province, China
| | - Huan He
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, Guangdong Province, China
| | - Xupeng Yue
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, Guangdong Province, China
| |
Collapse
|
9
|
Zhao Z, Jin G, Yao K, Liu K, Liu F, Chen H, Wang K, Gorja DR, Reddy K, Bode AM, Guo Z, Dong Z. Aurora B kinase as a novel molecular target for inhibition the growth of osteosarcoma. Mol Carcinog 2019; 58:1056-1067. [PMID: 30790360 DOI: 10.1002/mc.22993] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 01/07/2023]
Abstract
Osteosarcoma is the primary human malignant tumor affecting bone. This cancer most frequently arises in children and adolescents, with a second peak in those over the age of 50. Currently, surgery followed by radiotherapy and chemotherapy are the main treatments, but long-term positive effects are very poor. Aurora B kinase is a serine/threonine kinase that is a key regulator of cell cycle and mitosis. Tissue array analysis revealed that Aurora B kinase is overexpressed in osteosarcoma compared with normal bone tissue. We developed a compound, HOI-07 (i.e., (E)-3-((E)-4-(benzo[d] [1,3]dioxol-5-yl)-2-oxobut-3-en-1-ylidene)indolin-2-one), as a specific Aurora B kinase inhibitor and examined its effectiveness against osteosarcoma cell growth in this study. This compound inhibited Aurora B kinase activity in osteosarcoma and induced apoptosis, caused G2-M phase arrest, and attenuated osteosarcoma anchorage-independent cell growth. Moreover, knocking down the expression of Aurora B effectively reduced the sensitivity of osteosarcoma to HOI-07. Results of a xenograft mouse study indicated that HOI-07 treatment effectively suppressed the growth of 143B and KHOS xenografts, without affecting the body weight of mice. The expression of phosphorylated histone H3 (Ser10) was reduced in mice treated with HOI-07. Overall, we identified HOI-07 as a specific Aurora B kinase inhibitor for osteosarcoma treatment and this compound warrants further investigation.
Collapse
Affiliation(s)
- Zhenjiang Zhao
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,Henan Provincial Orthopedic Hospital, Zhengzhou, Henan, People's Republic of China
| | - Guoguo Jin
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,Henan Provincial Orthopedic Hospital, Zhengzhou, Henan, People's Republic of China
| | - Ke Yao
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,Henan Provincial Orthopedic Hospital, Zhengzhou, Henan, People's Republic of China
| | - Kangdong Liu
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, People's Republic of China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, People's Republic of China
| | - Fangfang Liu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, People's Republic of China
| | - Hanyong Chen
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,Henan Provincial Orthopedic Hospital, Zhengzhou, Henan, People's Republic of China
| | - Keke Wang
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, People's Republic of China
| | - Dhilli Rao Gorja
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, People's Republic of China
| | - Kanamata Reddy
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Zhiping Guo
- Henan Provincial Orthopedic Hospital, Zhengzhou, Henan, People's Republic of China
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
10
|
Wang W, Feng X, Liu HX, Chen SW, Hui L. Synthesis and biological evaluation of 2,4-disubstituted phthalazinones as Aurora kinase inhibitors. Bioorg Med Chem 2018; 26:3217-3226. [DOI: 10.1016/j.bmc.2018.04.048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/20/2018] [Accepted: 04/21/2018] [Indexed: 02/06/2023]
|
11
|
Cui S, Zhang K, Li C, Chen J, Pan Y, Feng B, Lu L, Zhu Z, Wang R, Chen L. Methylation-associated silencing of microRNA-129-3p promotes epithelial-mesenchymal transition, invasion and metastasis of hepatocelluar cancer by targeting Aurora-A. Oncotarget 2018; 7:78009-78028. [PMID: 27793005 PMCID: PMC5363640 DOI: 10.18632/oncotarget.12870] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/14/2016] [Indexed: 12/12/2022] Open
Abstract
Metastasis and recurrence has become one major obstacle for further improving the survival of hepatocelluar cancer (HCC) patients. Therefore, it is critical to elucidate the mechanisms involved in HCC metastasis. This study aimed to investigate the roles of microRNA (miR)-129-3p in HCC metastasis and its possible molecular mechanisms. By using microarray analysis to compare levels of different miRNAs in HCC tissues with or without lymph node metastasis (LNM), we showed that HCC tissues with LNM had reduced levels of miR-129-3p, which was related to its promoter hypermethylation and correlated with tumor metastasis, recurrence and poor prognosis. Gain - and loss - of - function assays indicated that re-expression of miR-129-3p could reverse epithelial-mesenchymal transition (EMT), and reduce in vitro invasion and in vivo metastasis of HCC cells. Aurora-A, a serine/threonine protein kinase, was identified as a direct target of miR-129-3p. Knockdown of Aurora-A phenocopied the effect of miR-129-3p overexpression on HCC metastasis. In addition, Aurora-A upregulation could partially rescue the effect of miR-129-3p. We further demonstrated that activation of PI3K/Akt and p38-MAPK signalings were involved in miR-129-3p-mediated HCC metastasis. These findings suggest that methylation-mediated miR-129-3p downregulation promotes EMT, in vitro invasion and in vivo metastasis of HCC cells via activation of PI3K/Akt and p38-MAPK signalings partially by targeting Aurora-A. Therefore, miR-129-3p may be a novel prognostic biomarker and potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Shiyun Cui
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| | - Kai Zhang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| | - Chen Li
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| | - Jing Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| | - Yan Pan
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| | - Bing Feng
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| | - Lei Lu
- Liver Disease Center of PLA, The 81th Hospital of PLA, Nanjing 210002, Jiangsu, PR China
| | - Ziman Zhu
- Department of Hepatobiliary Surgery, First Hospital Affiliated to the Chinese PLA General Hospital, Haidian District, Beijing 100048, PR China
| | - Rui Wang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| | - Longbang Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| |
Collapse
|
12
|
Lykkesfeldt AE, Iversen BR, Jensen MB, Ejlertsen B, Giobbie-Hurder A, Reiter BE, Kirkegaard T, Rasmussen BB. Aurora kinase A as a possible marker for endocrine resistance in early estrogen receptor positive breast cancer. Acta Oncol 2018; 57:67-73. [PMID: 29202611 DOI: 10.1080/0284186x.2017.1404126] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Cell culture studies have disclosed that the mitotic Aurora kinase A is causally involved in both tamoxifen and aromatase inhibitor resistant cell growth and thus may be a potential new marker for endocrine resistance in the clinical setting. MATERIAL AND METHODS Archival tumor tissue was available from 1323 Danish patients with estrogen receptor (ER) positive primary breast cancer, who participated in the Breast International Group (BIG) 1-98 trial, comparing treatment with tamoxifen and letrozole and both in a sequence. The expression of Aurora A was determined by immunohistochemistry in 980 tumors and semi quantitively scored into three groups; negative/weak, moderate and high. The Aurora A expression levels were compared to other clinico-pathological parameters and outcome, defined as disease-free survival (DFS) and overall survival (OS). RESULTS High expression of Aurora A was found in 26.9% of patients and moderate in 57.0%. High expression was significantly associated with high malignancy grade and HER2 amplification. High Aurora A expression was significantly more frequent in ductal compared to lobular carcinomas. We found no significant association between Aurora A expression and DFS or OS and no evidence of interaction between Aurora A expression and benefits from tamoxifen versus letrozole. CONCLUSIONS Aurora A expression in breast tumors was associated with high malignancy grade III and with HER2 amplification. A trend as a prognostic factor for OS was found in patients with high Aurora A expression. No predictive property was observed in this study with early breast cancer.
Collapse
Affiliation(s)
- Anne E. Lykkesfeldt
- Unit of Cell Death and Metabolism, Danish Cancer Society Research Center, Copenhagen, Denmark
| | | | - Maj-Britt Jensen
- Danish Breast Cancer Cooperative Group (DBCG), Rigshospitalet, Copenhagen, Denmark
| | - Bent Ejlertsen
- Danish Breast Cancer Cooperative Group (DBCG), Rigshospitalet, Copenhagen, Denmark
| | - Anita Giobbie-Hurder
- International Breast Cancer Study Group (IBCSG), Department of Biostatistics & Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Birgit E. Reiter
- Unit of Cell Death and Metabolism, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Tove Kirkegaard
- Department of Surgery, Zealand University Hospital, Koege, Denmark
| | | |
Collapse
|
13
|
Qin WW, Sang CY, Zhang LL, Wei W, Tian HZ, Liu HX, Chen SW, Hui L. Synthesis and biological evaluation of 2,4-diaminopyrimidines as selective Aurora A kinase inhibitors. Eur J Med Chem 2015; 95:174-84. [DOI: 10.1016/j.ejmech.2015.03.044] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 03/17/2015] [Accepted: 03/19/2015] [Indexed: 12/25/2022]
|
14
|
Abstract
For the past 30 years, improvements in the survival of patients with osteosarcoma have been mostly incremental. Despite evidence of genomic instability and a high frequency of chromothripsis and kataegis, osteosarcomas carry few recurrent targetable mutations, and trials of targeted agents have been generally disappointing. Bone has a highly specialized immune environment and many immune signalling pathways are important in bone homeostasis. The success of the innate immune stimulant mifamurtide in the adjuvant treatment of non-metastatic osteosarcoma suggests that newer immune-based treatments, such as immune checkpoint inhibitors, may substantially improve disease outcome.
Collapse
Affiliation(s)
- Maya Kansara
- 1] Research Division, Peter MacCallum Cancer Centre, Melbourne, 3002, Victoria, Australia. [2] Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, 3010, Victoria, Australia
| | - Michele W Teng
- 1] Immunology in Cancer and Infection Laboratory and Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, 4006, Queensland, Australia. [2] School of Medicine, University of Queensland, Herston, 4006, Queensland, Australia
| | - Mark J Smyth
- 1] Immunology in Cancer and Infection Laboratory and Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, 4006, Queensland, Australia. [2] School of Medicine, University of Queensland, Herston, 4006, Queensland, Australia
| | - David M Thomas
- 1] Research Division, Peter MacCallum Cancer Centre, Melbourne, 3002, Victoria, Australia. [2] Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, 3010, Victoria, Australia. [3] The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, 2010, New South Wales, Australia
| |
Collapse
|
15
|
Vassilopoulos A, Tominaga Y, Kim HS, Lahusen T, Li B, Yu H, Gius D, Deng CX. WEE1 murine deficiency induces hyper-activation of APC/C and results in genomic instability and carcinogenesis. Oncogene 2014; 34:3023-35. [PMID: 25088202 DOI: 10.1038/onc.2014.239] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 06/19/2014] [Accepted: 06/26/2014] [Indexed: 12/23/2022]
Abstract
The tyrosine kinase WEE1 controls the timing of entry into mitosis in eukaryotes and its genetic deletion leads to pre-implantation lethality in mice. Here, we show that besides the premature mitotic entry phenotype, Wee1 mutant murine cells fail to complete mitosis properly and exhibit several additional defects that contribute to the deregulation of mitosis, allowing mutant cells to progress through mitosis at the expense of genomic integrity. WEE1 interacts with the anaphase promoting complex, functioning as a negative regulator, and the deletion of Wee1 results in hyper-activation of this complex. Mammary specific knockout mice overcome the DNA damage response pathway triggered by the mis-coordination of the cell cycle in mammary epithelial cells and heterozygote mice spontaneously develop mammary tumors. Thus, WEE1 functions as a haploinsufficient tumor suppressor that coordinates distinct cell division events to allow correct segregation of genetic information into daughter cells and maintain genome integrity.
Collapse
Affiliation(s)
- A Vassilopoulos
- 1] Genetics of Development and Disease Branch, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA [2] Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Y Tominaga
- Genetics of Development and Disease Branch, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - H-Seok Kim
- 1] Genetics of Development and Disease Branch, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA [2] Department of Life Science, Ewha Womans University, Seoul, South Korea
| | - T Lahusen
- Genetics of Development and Disease Branch, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - B Li
- Department of Pharmacology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - H Yu
- Department of Pharmacology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - D Gius
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - C-X Deng
- Genetics of Development and Disease Branch, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
16
|
Cassol CA, Winer D, Liu W, Guo M, Ezzat S, Asa SL. Tyrosine kinase receptors as molecular targets in pheochromocytomas and paragangliomas. Mod Pathol 2014; 27:1050-62. [PMID: 24390213 PMCID: PMC4977182 DOI: 10.1038/modpathol.2013.233] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 10/15/2013] [Accepted: 10/16/2013] [Indexed: 12/17/2022]
Abstract
Pheochromocytomas and paragangliomas are neuroendocrine tumors shown to be responsive to multitargeted tyrosine kinase inhibitor (TKI) treatment. Despite growing knowledge regarding their genetic basis, the ability to predict behavior in these tumors remains challenging. There is also limited knowledge of their tyrosine kinase receptor expression and whether the clinical response observed to the TKI sunitinib relates only to its anti-angiogenic properties or also due to a direct effect on tumor cells. To answer these questions, an in vitro model of sunitinib treatment of a pheochromocytoma cell line was created. Sunitinib targets (VEGFRs, PDGFRs, and C-KIT), FGFRs, and cell cycle regulatory proteins were investigated in human tissue microarrays. SDHB immunohistochemistry was used as a surrogate marker for the presence of succinate dehydrogenase mutations. The FGFR4 G388R single nucleotide polymorphism was also investigated. Sunitinib treatment in vitro decreases cell proliferation mainly by targeting cell cycle, DNA metabolism, and cell organization genes. FGFR1, -2, and -4, VEGFR2, PDGFRα, and p16 were overexpressed in primary human pheochromocytomas and paragangliomas. Discordant results were observed for VEGFR1, p27, and p21 overexpressed in paragangliomas but underexpressed in pheochromocytomas; PDGFRβ, Rb, and Cyclin D1 overexpressed in paragangliomas only; and FGFR3 overexpressed in pheochromocytomas and underexpressed in paragangliomas. Low expression of C-KIT, p53, and Aurora kinase A and B was observed. Nuclear FGFR2 expression was associated with increased risk of metastasis (odds ratio (OR)=7.61, P=0.008), as was membranous PDGFRα (OR=13.71, P=0.015), membranous VEGFR1 (OR=8.01, P=0.037), nuclear MIB1 (OR=1.26, P=0.008), and cytoplasmic p27 (OR=1.037, P=0.030). FGFR3, VEGFR2, and C-KIT levels were associated with decreased risk of metastasis. We provide new insights into the mechanistic actions of sunitinib in pheochromocytomas and paragangliomas, and support current evidence that multitargeted TKIs might be a suitable treatment alternative for these tumors.
Collapse
Affiliation(s)
- Clarissa A. Cassol
- Department of Pathology, University Health Network, Toronto, Ontario, Canada
| | - Daniel Winer
- Department of Pathology, University Health Network, Toronto, Ontario, Canada
| | - Wei Liu
- Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | - Miao Guo
- Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | - Shereen Ezzat
- Department of Internal Medicine, University Health Network, Toronto, Ontario, Canada,Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | - Sylvia L. Asa
- Department of Pathology, University Health Network, Toronto, Ontario, Canada,Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Tavanti E, Sero V, Vella S, Fanelli M, Michelacci F, Landuzzi L, Magagnoli G, Versteeg R, Picci P, Hattinger CM, Serra M. Preclinical validation of Aurora kinases-targeting drugs in osteosarcoma. Br J Cancer 2013; 109:2607-18. [PMID: 24129234 PMCID: PMC3833226 DOI: 10.1038/bjc.2013.643] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 09/20/2013] [Accepted: 09/24/2013] [Indexed: 12/19/2022] Open
Abstract
Background: Aurora kinases are key regulators of cell cycle and represent new promising therapeutic targets in several human tumours. Methods: Biological relevance of Aurora kinase-A and -B was assessed on osteosarcoma clinical samples and by silencing these genes with specific siRNA in three human osteosarcoma cell lines. In vitro efficacy of two Aurora kinases-targeting drugs (VX-680 and ZM447439) was evaluated on a panel of four drug-sensitive and six drug-resistant human osteosarcoma cell lines. Results: Human osteosarcoma cell lines proved to be highly sensitive to both drugs. A decreased drug sensitivity was observed in doxorubicin-resistant cell lines, most probably related to ABCB1/MDR1 overexpression. Both drugs variably induced hyperploidy and apoptosis in the majority of cell lines. VX-680 also reduced in vitro cell motility and soft-agar cloning efficiency. Drug association experiments showed that VX-680 positively interacts with all conventional drugs used in osteosarcoma chemotherapy, overcoming the cross-resistance observed in the single-drug treatments. Conclusion: Aurora kinase-A and -B represent new candidate therapeutic targets for osteosarcoma. In vitro analysis of the Aurora kinases inhibitors VX-680 and ZM447439 indicated in VX-680 a new promising drug of potential clinical usefulness in association with conventional osteosarcoma chemotherapeutic agents.
Collapse
Affiliation(s)
- E Tavanti
- Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Via di Barbiano 1/10, I-40136 Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Cui SY, Huang JY, Chen YT, Song HZ, Huang GC, De W, Wang R, Chen LB. The role of Aurora A in hypoxia-inducible factor 1α-promoting malignant phenotypes of hepatocelluar carcinoma. Cell Cycle 2013; 12:2849-66. [PMID: 23966163 DOI: 10.4161/cc.25916] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Overexpression of both hypoxia-inducible factor 1α (HIF-1α) and Aurora A has been found in hepatocellular carcinoma (HCC). However, whether HIF-1α and Aurora A synergistically promote malignant phenotypes of HCC cells is unknown. The purpose of this study was to investigate the roles and functional correlation of HIF-1α and Aurora A in HCC progression. Immunohistochemistry was performed to detect HIF-1α and Aurora A protein expression in 55 primary HCC and corresponding non-tumor tissues and their clinical significance. Gene knockout technology using short hairpin RNA (shRNA) was used to knockdown expression of HIF-1α or Aurora A and analyze their effects on malignant phenotypes of HCC cells. The transcriptional regulation of Aurora A by HIF-1α and the possible downstream molecular signaling pathways were also determined. Results showed that hypoxia could induce the increased expression of HIF-1α and Aurora A in HCC cells. Also, shRNA-mediated HIF-1α downregulation could lead to the decreased Aurora A expression and inhibition of growth or invasion in HCC cells. Moreover, HIF-1α could transcriptionally regulate Aurora A expression by binding to hypoxia-responsive elements in the Aurora A promoter and recruiting the coactivator-p300/CBP. Additionally, shRNA-mediated Aurora A knockdown could mimic the effects of HIF-1α downregulation on phenotypes of HCC cells, and overexpression of Aurora A could partially rescue the phenotypical changes of HCC cells induced by HIF-1α downregulation. Further research indicated that activation of Akt and p38-MAPK signaling pathways mediated the downstream effects of HIF-1α and Aurora A in HCC cells under hypoxic condition. Taken together, our findings indicated that Aurora A might be a key regulator of HIF-1α-promoting malignant phenotypes of HCC by activation of Akt and p38-MAPK signaling pathways.
Collapse
Affiliation(s)
- Shi-Yun Cui
- Department of Medical Oncology; Jinling Hospital; School of Medicine; Nanjing University; Nanjing, PR China
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Evans MS, Madhunapantula SV, Robertson GP, Drabick JJ. Current and future trials of targeted therapies in cutaneous melanoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 779:223-55. [PMID: 23288642 DOI: 10.1007/978-1-4614-6176-0_10] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In order to effectively treat melanoma, targeted inhibition of key m-echanistic events regulating melanoma development such as cell proliferation, survival, angiogenesis and invasion or metastasis needs to be accomplished. The Mitogen Activated Protein Kinase (MAPK) pathway has been identified as a key player in melanoma development making this cascade an important therapeutic target. However, identification of the ideal pathway member to therapeutically target for maximal clinical benefit remains a challenge. In normal cells, the MAPK pathway relays extracellular signals from the cell membrane to the nucleus via a cascade of phosphorylation events, which promote cancer development. Dysregulation of the MAPK pathway occurs frequently in many human cancers including melanoma. Mutations in the B-RAF and RAS genes, genetic or epigenetic modifications are the key aberrations observed in this signaling cascade. Constitutive activation of this pathway causes oncogenic transformation of cells by promoting cell proliferation, invasion, metastasis, migration, survival and angiogenesis. This review provides an overview of (a) key members of MAPK signaling regulating melanoma development; (b) key proteins which can serve as biomarkers to assess disease progression; (c) the clinical efficacy of various pharmacological agents targeting MAPK pathway; (d) current clinical trials evaluating downstream targets of the MAPK pathway; (e) issues associated with pharmacological agents such as drug resistance, induction of cancers; and finally (e) various strategies overcoming drug resistance.
Collapse
Affiliation(s)
- Matthew S Evans
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | | | | | | |
Collapse
|
20
|
Li L, Yang G, Ren C, Tanimoto R, Hirayama T, Wang J, Hawke D, Kim SM, Lee JS, Goltsov AA, Park S, Ittmann MM, Troncoso P, Thompson TC. Glioma pathogenesis-related protein 1 induces prostate cancer cell death through Hsc70-mediated suppression of AURKA and TPX2. Mol Oncol 2012; 7:484-96. [PMID: 23333597 DOI: 10.1016/j.molonc.2012.12.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 12/04/2012] [Accepted: 12/20/2012] [Indexed: 01/09/2023] Open
Abstract
In this study we report that expression of glioma pathogenesis-related protein 1 (GLIPR1) regulated numerous apoptotic, cell cycle, and spindle/centrosome assembly-related genes, including AURKA and TPX2, and induced apoptosis and/or mitotic catastrophe (MC) in prostate cancer (PCa) cells, including p53-mutated/deleted, androgen-insensitive metastatic PCa cells. Mechanistically, GLIPR1 interacts with heat shock cognate protein 70 (Hsc70); this interaction is associated with SP1 and c-Myb destabilization and suppression of SP1- and c-Myb-mediated AURKA and TPX2 transcription. Inhibition of AURKA and TPX2 using siRNA mimicked enforced GLIPR1 expression in the induction of apoptosis and MC. Recombinant GLIPR1-ΔTM protein inhibited AURKA and TPX2 expression, induced apoptosis and MC, and suppressed orthotopic xenograft tumor growth. Our results define a novel GLIPR1-regulated signaling pathway that controls apoptosis and/or mitotic catastrophe in PCa cells and establishes the potential of this pathway for targeted therapies.
Collapse
Affiliation(s)
- Likun Li
- Department of Genitourinary Medical Oncology, Unit 18-3, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lawrence HR, Martin MP, Luo Y, Pireddu R, Yang H, Gevariya H, Ozcan S, Zhu JY, Kendig R, Rodriguez M, Elias R, Cheng JQ, Sebti SM, Schonbrunn E, Lawrence NJ. Development of o-chlorophenyl substituted pyrimidines as exceptionally potent aurora kinase inhibitors. J Med Chem 2012; 55:7392-7416. [PMID: 22803810 DOI: 10.1021/jm300334d] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The o-carboxylic acid substituted bisanilinopyrimidine 1 was identified as a potent hit (Aurora A IC(50) = 6.1 ± 1.0 nM) from in-house screening. Detailed structure-activity relationship (SAR) studies indicated that polar substituents at the para position of the B-ring are critical for potent activity. X-ray crystallography studies revealed that compound 1 is a type I inhibitor that binds the Aurora kinase active site in a DFG-in conformation. Structure-activity guided replacement of the A-ring carboxylic acid with halogens and incorporation of fluorine at the pyrimidine 5-position led to highly potent inhibitors of Aurora A that bind in a DFG-out conformation. B-Ring modifications were undertaken to improve the solubility and cell permeability. Compounds such as 9m with water-solubilizing moieties at the para position of the B-ring inhibited the autophosphorylation of Aurora A in MDA-MB-468 breast cancer cells.
Collapse
Affiliation(s)
- Harshani R Lawrence
- Department of Drug Discovery, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA.,Department of Chemical Biology Core, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA.,Department of Oncologic Sciences, University of South Florida, Tampa, FL 33620, USA
| | - Matthew P Martin
- Department of Drug Discovery, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Yunting Luo
- Department of Chemical Biology Core, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Roberta Pireddu
- Department of Drug Discovery, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Hua Yang
- Department of Drug Discovery, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Harsukh Gevariya
- Department of Drug Discovery, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Sevil Ozcan
- Department of Drug Discovery, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Jin-Yi Zhu
- Department of Drug Discovery, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Robert Kendig
- Department of Chemical Biology Core, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Mercedes Rodriguez
- Department of Chemical Biology Core, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA.,Department of Drug Discovery, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Roy Elias
- Department of Drug Discovery, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Jin Q Cheng
- Department of Molecular Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA.,Department of Oncologic Sciences, University of South Florida, Tampa, FL 33620, USA
| | - Saïd M Sebti
- Department of Drug Discovery, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA.,Department of Oncologic Sciences, University of South Florida, Tampa, FL 33620, USA.,Department of Molecular Medicine, University of South Florida, Tampa, FL 33620, USA
| | - Ernst Schonbrunn
- Department of Drug Discovery, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA.,Department of Chemical Biology Core, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA.,Department of Oncologic Sciences, University of South Florida, Tampa, FL 33620, USA
| | - Nicholas J Lawrence
- Department of Drug Discovery, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA.,Department of Oncologic Sciences, University of South Florida, Tampa, FL 33620, USA
| |
Collapse
|
22
|
Yang S, Ma J, Xiao J, Lv X, Li X, Yang H, Liu Y, Feng S, Zhang Y. Arctigenin anti-tumor activity in bladder cancer T24 cell line through induction of cell-cycle arrest and apoptosis. Anat Rec (Hoboken) 2012; 295:1260-6. [PMID: 22619087 DOI: 10.1002/ar.22497] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Revised: 03/26/2012] [Accepted: 04/16/2012] [Indexed: 01/15/2023]
Abstract
Bladder cancer is the most common neoplasm in the urinary system. This study assesses arctigenin anti-tumor activity in human bladder cancer T24 cells in vitro and the underlying molecular events. The flow cytometry analysis was used to detect cell-cycle distribution and apoptosis. Western blotting was used to detect changes in protein expression. The data showed that arctigenin treatment reduced viability of bladder cancer T24 cells in a dose- and time-dependent manner after treatment with arctigenin (10, 20, 40, 80, and 100 μmol/L) for 24 hr and 48 hr. Arctigenin treatment clearly arrested tumor cells in the G1 phase of the cell cycle. Apoptosis was detected by hoechst stain and flow cytometry after Annexin-V-FITC/PI double staining. Early and late apoptotic cells were accounted for 2.32-7.01% and 3.07-7.35%, respectively. At the molecular level, arctigenin treatment decreased cyclin D1 expression, whereas CDK4 and CDK6 expression levels were unaffected. Moreover, arctigenin selectively altered the phosphorylation of members of the MAPK superfamily, decreasing phosphorylation of ERK1/2 and activated phosphorylation of p38 significantly in a dose-dependent manner. These results suggest that arctigenin may inhibit cell viability and induce apoptosis by direct activation of the mitochondrial pathway, and the mitogen-activated protein kinase pathway may play an important role in the anti-tumor effect of arctigenin. The data from the current study demonstrate the usefulness of arctigenin in bladder cancer T24 cells, which should further be evaluated in vivo before translation into clinical trials for the chemoprevention of bladder cancer.
Collapse
Affiliation(s)
- Shucai Yang
- Department of Anatomy, Harbin Medical University, Harbin, China
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Wang JX, Zeng Q, Chen L, Du JC, Yan XL, Yuan HF, Zhai C, Zhou JN, Jia YL, Yue W, Pei XT. SPINDLIN1 Promotes Cancer Cell Proliferation through Activation of WNT/TCF-4 Signaling. Mol Cancer Res 2012; 10:326-35. [DOI: 10.1158/1541-7786.mcr-11-0440] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
24
|
Cyclin B1 expression regulated by cytoplasmic polyadenylation element binding protein in astrocytes. J Neurosci 2011; 31:12118-28. [PMID: 21865454 DOI: 10.1523/jneurosci.1621-11.2011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Astrocytes are the most abundant cells in the brain, playing vital roles in neuronal survival, growth, and function. Understanding the mechanism(s) regulating astrocyte proliferation will have important implications in brain development, response to injury, and tumorigenesis. Cyclin B1 is well known to be a critical regulator of mitotic entry via its interaction with cyclin-dependent kinase 1. In rat astrocytes, we now show that the mRNA binding protein cytoplasmic polyadenylation element binding protein 1 (CPEB1) is associated with cyclin B1 mRNA and that this interaction is enriched at the centrosome. In addition, if growth-arrested astrocytes are stimulated to divide, CPEB1 is phosphorylated and cyclin B1 mRNA is polyadenylated, both hallmarks of CPEB1 activation, resulting in an increase in cyclin B1 protein. CPEB1 binding to mRNA initially inhibits translation; therefore, removing CPEB1 from mRNA should result in an increase in translation due to derepression. Indeed, when we either knocked down CPEB1 protein with siRNA or sequestered it from endogenous mRNA by expressing RNA containing multiple CPEB1 binding sites, cyclin B1 protein was increased and cell proliferation was stimulated. Our data suggest a mechanism wherein CPEB1 is bound and represses cyclin B1 mRNA translation until a signal to proliferate phosphorylates CPEB1, resulting in an increase in cyclin B1 protein and progression into mitosis. Our results demonstrate for the first time a role for CPEB1 in regulating cell proliferation in the brain.
Collapse
|
25
|
Kim HS, Vassilopoulos A, Wang RH, Lahusen T, Xiao Z, Xu X, Li C, Veenstra TD, Li B, Yu H, Ji J, Wang XW, Park SH, Cha YI, Gius D, Deng CX. SIRT2 maintains genome integrity and suppresses tumorigenesis through regulating APC/C activity. Cancer Cell 2011; 20:487-99. [PMID: 22014574 PMCID: PMC3199577 DOI: 10.1016/j.ccr.2011.09.004] [Citation(s) in RCA: 414] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 05/26/2011] [Accepted: 09/08/2011] [Indexed: 11/17/2022]
Abstract
Members of sirtuin family regulate multiple critical biological processes, yet their role in carcinogenesis remains controversial. To investigate the physiological functions of SIRT2 in development and tumorigenesis, we disrupted Sirt2 in mice. We demonstrated that SIRT2 regulates the anaphase-promoting complex/cyclosome activity through deacetylation of its coactivators, APC(CDH1) and CDC20. SIRT2 deficiency caused increased levels of mitotic regulators, including Aurora-A and -B that direct centrosome amplification, aneuploidy, and mitotic cell death. Sirt2-deficient mice develop gender-specific tumorigenesis, with females primarily developing mammary tumors, and males developing more hepatocellular carcinoma (HCC). Human breast cancers and HCC samples exhibited reduced SIRT2 levels compared with normal tissues. These data demonstrate that SIRT2 is a tumor suppressor through its role in regulating mitosis and genome integrity.
Collapse
Affiliation(s)
- Hyun-Seok Kim
- Genetics of Development and Disease Branch, 10/9N105, National Institute of Diabetes, Digestive and Kidney Diseases; National Institutes of Health, Bethesda, MD 20892, USA
- Department of Radiation Oncology and Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Athanassios Vassilopoulos
- Genetics of Development and Disease Branch, 10/9N105, National Institute of Diabetes, Digestive and Kidney Diseases; National Institutes of Health, Bethesda, MD 20892, USA
| | - Rui-Hong Wang
- Genetics of Development and Disease Branch, 10/9N105, National Institute of Diabetes, Digestive and Kidney Diseases; National Institutes of Health, Bethesda, MD 20892, USA
| | - Tyler Lahusen
- Genetics of Development and Disease Branch, 10/9N105, National Institute of Diabetes, Digestive and Kidney Diseases; National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhen Xiao
- Laboratory of Proteomics and Analytical Technologies, Advanced Technology Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD, 20170, USA
| | - Xiaoling Xu
- Genetics of Development and Disease Branch, 10/9N105, National Institute of Diabetes, Digestive and Kidney Diseases; National Institutes of Health, Bethesda, MD 20892, USA
| | - Cuiling Li
- Genetics of Development and Disease Branch, 10/9N105, National Institute of Diabetes, Digestive and Kidney Diseases; National Institutes of Health, Bethesda, MD 20892, USA
| | - Timothy D. Veenstra
- Laboratory of Proteomics and Analytical Technologies, Advanced Technology Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD, 20170, USA
| | - Bing Li
- Department of Pharmacology, UT Southwestern Medical Center, 6001 Forest Park Rd, Dallas, TX 75390, USA
| | - Hongtao Yu
- Department of Pharmacology, UT Southwestern Medical Center, 6001 Forest Park Rd, Dallas, TX 75390, USA
| | - Junfang Ji
- Laboratory of Human Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Seong-Hoon Park
- Department of Radiation Oncology and Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Yong I. Cha
- Department of Radiation Oncology and Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - David Gius
- Department of Radiation Oncology and Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Chu-Xia Deng
- Genetics of Development and Disease Branch, 10/9N105, National Institute of Diabetes, Digestive and Kidney Diseases; National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
26
|
Corrionero A, Miñana B, Valcárcel J. Reduced fidelity of branch point recognition and alternative splicing induced by the anti-tumor drug spliceostatin A. Genes Dev 2011; 25:445-59. [PMID: 21363963 DOI: 10.1101/gad.2014311] [Citation(s) in RCA: 205] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Spliceostatin A (SSA) is a stabilized derivative of a Pseudomonas bacterial fermentation product that displays potent anti-proliferative and anti-tumor activities in cancer cells and animal models. The drug inhibits pre-mRNA splicing in vitro and in vivo and binds SF3b, a protein subcomplex of U2 small nuclear ribonucleoprotein (snRNP), which is essential for recognition of the pre-mRNA branch point. We report that SSA prevents interaction of an SF3b 155-kDa subunit with the pre-mRNA, concomitant with nonproductive recruitment of U2 snRNP to sequences 5' of the branch point. Differences in base-pairing potential with U2 snRNA in this region lead to different sensitivity of 3' splice sites to SSA, and to SSA-induced changes in alternative splicing. Indeed, rather than general splicing inhibition, splicing-sensitive microarray analyses reveal specific alternative splicing changes induced by the drug that significantly overlap with those induced by knockdown of SF3b 155. These changes lead to down-regulation of genes important for cell division, including cyclin A2 and Aurora A kinase, thus providing an explanation for the anti-proliferative effects of SSA. Our results reveal a mechanism that prevents nonproductive base-pairing interactions in the spliceosome, and highlight the regulatory and cancer therapeutic potential of perturbing the fidelity of splice site recognition.
Collapse
|
27
|
Wu L, Shao L, An N, Wang J, Pazhanisamy S, Feng W, Hauer-Jensen M, Miyamoto S, Zhou D. IKKβ regulates the repair of DNA double-strand breaks induced by ionizing radiation in MCF-7 breast cancer cells. PLoS One 2011; 6:e18447. [PMID: 21490922 PMCID: PMC3072401 DOI: 10.1371/journal.pone.0018447] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 02/28/2011] [Indexed: 01/17/2023] Open
Abstract
Activation of the IKK-NFκB pathway increases the resistance of cancer cells to ionizing radiation (IR). This effect has been largely attributed to the induction of anti-apoptotic proteins by NFκB. Since efficient repair of DNA double strand breaks (DSBs) is required for the clonogenic survival of irradiated cells, we investigated if activation of the IKK-NFκB pathway also regulates DSB repair to promote cell survival after IR. We found that inhibition of the IKK-NFκB pathway with a specific IKKβ inhibitor significantly reduced the repair of IR-induced DSBs in MCF-7 cells. The repair of DSBs was also significantly inhibited by silencing IKKβ expression with IKKβ shRNA. However, down-regulation of IKKα expression with IKKα shRNA had no significant effect on the repair of IR-induced DSBs. Similar findings were also observed in IKKα and/or IKKβ knockout mouse embryonic fibroblasts (MEFs). More importantly, inhibition of IKKβ with an inhibitor or down-regulation of IKKβ with IKKβ shRNA sensitized MCF-7 cells to IR-induced clonogenic cell death. DSB repair function and resistance to IR were completely restored by IKKβ reconstitution in IKKβ-knockdown MCF-7 cells. These findings demonstrate that IKKβ can regulate the repair of DSBs, a previously undescribed and important IKKβ kinase function; and inhibition of DSB repair may contribute to cance cell radiosensitization induced by IKKβ inhibition. As such, specific inhibition of IKKβ may represents a more effective approach to sensitize cancer cells to radiotherapy.
Collapse
Affiliation(s)
- Lixian Wu
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Department of Pharmacology, Fujian Medical University, Fuzhou, China
| | - Lijian Shao
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Ningfei An
- Department of Pathology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Junru Wang
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Senthil Pazhanisamy
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Wei Feng
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, United States of America
| | - Shigeki Miyamoto
- Department of Pharmacology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Daohong Zhou
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- * E-mail:
| |
Collapse
|
28
|
Hattinger CM, Pasello M, Ferrari S, Picci P, Serra M. Emerging drugs for high-grade osteosarcoma. Expert Opin Emerg Drugs 2010; 15:615-34. [PMID: 20690888 DOI: 10.1517/14728214.2010.505603] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD Osteosarcoma (OS) is the most common primary malignant bone tumour in children and adolescents. This review focuses on the most promising therapeutic markers and drugs which may potentially be considered for innovative high-grade OS treatments. AREAS COVERED IN THIS REVIEW The list of drugs and compounds reviewed has been generated by taking into account those which target markers of potential clinical interest for high-grade OS and have been included in Phase I, II or III clinical trials. The literature search covers the last 40 years, starting from the first OS chemotherapy reports of the early 1970s. Particular relevance was given to reports and reviews on new targeted therapies of possible clinical usefulness for high-grade OS. WHAT THE READER WILL GAIN This review gives an updated overview of novel therapeutic approaches which have been or are going to be evaluated in Phase I/II/III clinical studies for high-grade OS. TAKE HOME MESSAGE On the basis of the information that has emerged so far, it can be predicted that in the next 5 - 10 years, new agents to be included in innovative treatment strategies for selected subgroups of high-grade OS patients may become available.
Collapse
Affiliation(s)
- Claudia Maria Hattinger
- Rizzoli Orthopaedic Institute, Pharmacogenomics and Pharmacogenetics Research Unit, Laboratory of Experimental Oncology, Via di Barbiano 1/10, 40136 Bologna, Italy
| | | | | | | | | |
Collapse
|