1
|
Mahaddalkar T, Banerjee A, Ketkar M, Thorat R, Gardi N, Dutt S. Aurora Kinase A and B inhibition abrogates 'Neosis', a non-mitotic cell division of GBM residual cells and prevents GBM recurrence. Oncogene 2025:10.1038/s41388-025-03372-6. [PMID: 40195468 DOI: 10.1038/s41388-025-03372-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 02/28/2025] [Accepted: 03/25/2025] [Indexed: 04/09/2025]
Abstract
Glioblastoma (GBM) has a dismal median survival of 15 months owing to therapy resistance and inevitable recurrence. Using our cellular models of GBM radiation resistance, we had shown that GBM recurrence is due to survival and proliferation of residual disease cells enriched in multinucleated giant cells (MNGCs). However, MNGC division mechanism remained elusive. Here, using live-cell imaging we found daughter cells emerge from MNGCs by cytoplasmic pinching. Lack of DNA condensation, absence of spindle poles and acto-myosin contractile ring in dividing-MNGCs confirmed non-mitotic division of MNGCs. Furthermore, MNGCs harboured DNA damage, senescence phenotype, repeated atypical division after radiation exposure, characteristics of unconventional division called 'Neosis'. Molecularly, WGCNA co-expression network analysis of RNA-Sequencing from parent, non-dividing MNGCs and dividing-MNGCs identified significantly high expression of aurora kinases (AurA and AurB) specifically in dividing-MNGCs. Pharmacological and genetic inhibition of aurora kinases abrogated MNGC neosis, preventing GBM recurrence in vitro and in vivo in an orthotopic GBM mouse model. Together, this study demonstrates that MNGCs divide by neosis, an atypical division mediated by AurA and AurB and identify aurora kinases as a potential molecular target to inhibit neosis and prevent GBM recurrence.
Collapse
Affiliation(s)
- Tejashree Mahaddalkar
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| | - Archisman Banerjee
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| | - Madhura Ketkar
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| | - Rahul Thorat
- Laboratory Animal Facility, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India
| | - Nilesh Gardi
- Department of Medical Oncology, Tata Memorial Centre, Navi Mumbai, India
| | - Shilpee Dutt
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India.
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India.
- Shilpee Dutt Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, India.
| |
Collapse
|
2
|
Lemay SE, Mougin M, Sauvaget M, El Kabbout R, Valasarajan C, Yamamoto K, Martineau S, Pelletier A, Bilodeau C, Grobs Y, Bourgeois A, Romanet C, Breuils-Bonnet S, Montesinos MS, Lu M, Chen H, Gilbert M, Théberge C, Potus F, Pullamsetti S, Provencher S, Bonnet S, Boucherat O. Unraveling AURKB as a potential therapeutic target in pulmonary hypertension using integrated transcriptomic analysis and pre-clinical studies. Cell Rep Med 2025; 6:101964. [PMID: 39933527 PMCID: PMC11866512 DOI: 10.1016/j.xcrm.2025.101964] [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: 05/02/2024] [Revised: 09/29/2024] [Accepted: 01/15/2025] [Indexed: 02/13/2025]
Abstract
Despite advances in treatment, the prognosis for patients with pulmonary arterial hypertension (PAH) remains dismal, highlighting the need for further therapeutic advances. By using RNA sequencing on pulmonary artery smooth muscle cells (PASMCs), functional enrichment, and connectivity map analyses, we identify Aurora kinase B (AURKB) as a candidate therapeutic target. We show that AURKB inhibition blocks cell cycle progression and reverses the gene signature of PAH-PASMCs. We also report that PAH-PASMCs that escape apoptosis acquire a senescence-associated secretory phenotype. In vivo, AURKB inhibition using barasertib improves hemodynamics in two preclinical models of established PAH by attenuating pulmonary vascular remodeling. A therapeutic effect is also observed in human precision-cut lung slices. Finally, we demonstrate that the combination of barasertib with a p21 attenuator is more effective in reducing vascular remodeling than either drug alone. These findings provide insight into strategies for therapeutic manipulation.
Collapse
MESH Headings
- Humans
- Animals
- Aurora Kinase B/antagonists & inhibitors
- Aurora Kinase B/metabolism
- Aurora Kinase B/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Pulmonary Artery/pathology
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Vascular Remodeling/drug effects
- Gene Expression Profiling
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/pathology
- Transcriptome/genetics
- Mice
- Male
- Apoptosis/drug effects
- Disease Models, Animal
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular
- Cyclin-Dependent Kinase Inhibitor p21/metabolism
Collapse
Affiliation(s)
- Sarah-Eve Lemay
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Manon Mougin
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Mélanie Sauvaget
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Reem El Kabbout
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Chanil Valasarajan
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Keiko Yamamoto
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Sandra Martineau
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Andréanne Pelletier
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Coralie Bilodeau
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Yann Grobs
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Alice Bourgeois
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Charlotte Romanet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Sandra Breuils-Bonnet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | | | - Min Lu
- Morphic Therapeutic, Inc, Waltham, MA, USA
| | | | - Mégan Gilbert
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Charlie Théberge
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - François Potus
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada; Department of Medicine, Laval University, Québec City, QC, Canada
| | - Soni Pullamsetti
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, German Center for Lung Research (DZL), Bad Nauheim, Germany; Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), DZL, Justus Liebig University, member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Steeve Provencher
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada; Department of Medicine, Laval University, Québec City, QC, Canada
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada; Department of Medicine, Laval University, Québec City, QC, Canada.
| | - Olivier Boucherat
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada; Department of Medicine, Laval University, Québec City, QC, Canada.
| |
Collapse
|
3
|
Chen H, Li M, Zhang Y, Song M, Que Y, Wang J, Sun F, Zhu J, Huang J, Liu J, Xu J, Lu S, Zhang Y. AURKB inhibition induces rhabdomyosarcoma apoptosis and ferroptosis through NPM1/SP1/ACSL5 axis. JCI Insight 2025; 10:e182429. [PMID: 39927464 PMCID: PMC11948576 DOI: 10.1172/jci.insight.182429] [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: 05/01/2024] [Accepted: 12/19/2024] [Indexed: 02/11/2025] Open
Abstract
Rhabdomyosarcoma (RMS) is one of the most common solid tumors in children and adolescents. Patients with relapsed/refractory RMS have limited treatment options, highlighting the urgency for the identification of novel therapeutic targets for RMS. In the present study, aurora kinase B (AURKB) was found to be highly expressed in RMS and associated with unfavorable prognosis of patients. Functional experiments indicated that inhibition of AURKB significantly reduced RMS cell proliferation, induced apoptosis and ferroptosis, and suppressed RMS growth in vivo. The highly expressed AURKB in RMS contributes to the apoptosis and ferroptosis resistance of tumor cells through the nucleophosmin 1 (NPM1)/Sp1 transcription factor (SP1)/acyl-CoA synthetase long-chain family member 5 (ACSL5) axis. Furthermore, inhibition of AURKB exerted an anti-RMS effect together with vincristine both in vitro and in vivo, with tolerable toxicity. The above findings provide insights we believe are new into the tumorigenesis of RMS, especially with regard to apoptosis or ferroptosis resistance, indicating that AURKB may be a potential target for clinical intervention in patients with RMS.
Collapse
Affiliation(s)
- Huimou Chen
- Department of Pediatric Oncology and
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Mengzhen Li
- Department of Pediatric Oncology and
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yu Zhang
- Department of Pediatric Oncology and
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mengjia Song
- Department of Pediatric Oncology and
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yi Que
- Department of Pediatric Oncology and
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Juan Wang
- Department of Pediatric Oncology and
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Feifei Sun
- Department of Pediatric Oncology and
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jia Zhu
- Department of Pediatric Oncology and
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Junting Huang
- Department of Pediatric Oncology and
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Juan Liu
- Department of Pediatric Oncology and
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiaqian Xu
- Department of Pediatric Oncology and
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Suying Lu
- Department of Pediatric Oncology and
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yizhuo Zhang
- Department of Pediatric Oncology and
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
4
|
Zhao Y, Zheng Z, Jin X, Liang S, Zhang C, Zhang M, Lang Y, Li P, Liu Z. Aurora kinase B inhibitor AZD1152: repurposing for treatment of lupus nephritis driven by the results of clinical trials. EBioMedicine 2025; 112:105553. [PMID: 39799765 PMCID: PMC11773216 DOI: 10.1016/j.ebiom.2024.105553] [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: 01/25/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Lupus nephritis (LN) is one of the most common and severe complications of systemic lupus erythematosus (SLE). Multitarget therapy (MT) achieves a 20% higher complete remission (CR) rate compared to conventional therapy in LN management. Intrigued by its excellent clinical efficacy, we aimed to develop a single-agent therapy with comparable efficacy to MT, offering a simplified treatment regimen. METHODS AZD1152, an Aurora kinase B (Aurkb) inhibitor, was identified through transcriptomic analyses and the L1000 CMap drug repurposing database. The therapeutic efficacy of AZD1152 was evaluated in MRL/lpr mice. Transcriptome sequencing and functional assays were performed to elucidate its mechanisms of action. Aurkb expression and its clinical relevance were assessed in lupus-prone mice and patients with LN. FINDINGS AZD1152 significantly attenuated systemic immune activation and renal injury in MRL/lpr mice, demonstrating efficacy comparable to MT regimens in animal studies. AZD1152 treatment modulated immune-inflammatory pathways in the kidney. Aurkb expression was upregulated in T cells infiltrating the renal interstitium in LN. Additionally, Aurkb expression levels positively correlated with the activity index (AI) and serum creatinine (Scr) in patients with LN. Mechanistic studies revealed that AZD1152 exerts therapeutic effects primarily by inhibiting T-cell proliferation. INTERPRETATION This study presents a drug development strategy that integrates clinically validated LN therapies with drug repurposing approaches. This strategy could accelerate drug development and clinical translation processes for LN. FUNDING A full list of funding sources can be found in the acknowledgements section.
Collapse
Affiliation(s)
- Yue Zhao
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, China
| | - Zuguo Zheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xuexiao Jin
- Institute of Immunology and Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Shaoshan Liang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, China
| | - Changming Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, China
| | - Mingchao Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, China
| | - Yue Lang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, China.
| |
Collapse
|
5
|
Hosea R, Hillary S, Naqvi S, Wu S, Kasim V. The two sides of chromosomal instability: drivers and brakes in cancer. Signal Transduct Target Ther 2024; 9:75. [PMID: 38553459 PMCID: PMC10980778 DOI: 10.1038/s41392-024-01767-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/18/2024] [Accepted: 02/06/2024] [Indexed: 04/02/2024] Open
Abstract
Chromosomal instability (CIN) is a hallmark of cancer and is associated with tumor cell malignancy. CIN triggers a chain reaction in cells leading to chromosomal abnormalities, including deviations from the normal chromosome number or structural changes in chromosomes. CIN arises from errors in DNA replication and chromosome segregation during cell division, leading to the formation of cells with abnormal number and/or structure of chromosomes. Errors in DNA replication result from abnormal replication licensing as well as replication stress, such as double-strand breaks and stalled replication forks; meanwhile, errors in chromosome segregation stem from defects in chromosome segregation machinery, including centrosome amplification, erroneous microtubule-kinetochore attachments, spindle assembly checkpoint, or defective sister chromatids cohesion. In normal cells, CIN is deleterious and is associated with DNA damage, proteotoxic stress, metabolic alteration, cell cycle arrest, and senescence. Paradoxically, despite these negative consequences, CIN is one of the hallmarks of cancer found in over 90% of solid tumors and in blood cancers. Furthermore, CIN could endow tumors with enhanced adaptation capabilities due to increased intratumor heterogeneity, thereby facilitating adaptive resistance to therapies; however, excessive CIN could induce tumor cells death, leading to the "just-right" model for CIN in tumors. Elucidating the complex nature of CIN is crucial for understanding the dynamics of tumorigenesis and for developing effective anti-tumor treatments. This review provides an overview of causes and consequences of CIN, as well as the paradox of CIN, a phenomenon that continues to perplex researchers. Finally, this review explores the potential of CIN-based anti-tumor therapy.
Collapse
Affiliation(s)
- Rendy Hosea
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Sharon Hillary
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Sumera Naqvi
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Shourong Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China.
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China.
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China.
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
6
|
Lakkaniga NR, Wang Z, Xiao Y, Kharbanda A, Lan L, Li HY. Revisiting Aurora Kinase B: A promising therapeutic target for cancer therapy. Med Res Rev 2024; 44:686-706. [PMID: 37983866 DOI: 10.1002/med.21994] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 03/28/2023] [Accepted: 10/29/2023] [Indexed: 11/22/2023]
Abstract
Cancer continues to be a major health concern globally, although the advent of targeted therapy has revolutionized treatment options. Aurora Kinase B is a serine-threonine kinase that has been explored as an oncology therapeutic target for more than two decades. Aurora Kinase B inhibitors show promising biological results in in-vitro and in-vivo experiments. However, there are no inhibitors approved yet for clinical use, primarily because of the side effects associated with Aurora B inhibitors. Several studies demonstrate that Aurora B inhibitors show excellent synergy with various chemotherapeutic agents, radiation therapy, and targeted therapies. This makes it an excellent choice as an adjuvant therapy to first-line therapies, which greatly improves the therapeutic window and side effect profile. Recent studies indicate the role of Aurora B in some deadly cancers with limited therapeutic options, like triple-negative breast cancer and glioblastoma. Herein, we review the latest developments in Aurora Kinase B targeted research, with emphasis on its potential as an adjuvant therapy and its role in some of the most difficult-to-treat cancers.
Collapse
Affiliation(s)
- Naga Rajiv Lakkaniga
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines), Dhanbad, India
| | - Zhengyu Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Yao Xiao
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Anupreet Kharbanda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Li Lan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
7
|
Zhang J, Ma J, Li Y, An Y, Du W, Yang Q, Huang M, Cai X. Overexpression of Aurora Kinase B Is Correlated with Diagnosis and Poor Prognosis in Hepatocellular Carcinoma. Int J Mol Sci 2024; 25:2199. [PMID: 38396874 PMCID: PMC10889672 DOI: 10.3390/ijms25042199] [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: 11/08/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Aurora kinase B (AURKB) overexpression promotes tumor initiation and development by participating in the cell cycle. In this study, we focused on the mechanism of AURKB in hepatocellular carcinoma (HCC) progression and on AURKB's value as a diagnostic and prognostic biomarker in HCC. We used data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) to analyze AURKB expression in HCC. We found that the expression levels of AURKB in HCC samples were higher than those in the corresponding control group. R packages were used to analyze RNA sequencing data to identify AURKB-related differentially expressed genes (DEGs), and these genes were found to be significantly enriched during the cell cycle. The biological function of AURKB was verified, and the results showed that cell proliferation was slowed down and cells were arrested in the G2/M phase when AURKB was knocked down. AURKB overexpression resulted in significant differences in clinical symptoms, such as the clinical T stage and pathological stage. Kaplan-Meier survival analysis, Cox regression analysis, and Receiver Operating Characteristic (ROC) curve analysis suggested that AURKB overexpression has good diagnostic and prognostic potential in HCC. Therefore, AURKB may be used as a potential target for the diagnosis and cure of HCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xuefei Cai
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, 1 Yixue Yuan Road, Chongqing 400016, China; (J.Z.); (J.M.); (Y.L.); (Y.A.); (W.D.); (Q.Y.); (M.H.)
| |
Collapse
|
8
|
Ledvin L, Gassaway BM, Tawil J, Urso O, Pizzo D, Welsh KA, Bolhuis DL, Fisher D, Bonni A, Gygi SP, Brown NG, Ferguson CJ. The anaphase-promoting complex controls a ubiquitination-phosphoprotein axis in chromatin during neurodevelopment. Dev Cell 2023; 58:2666-2683.e9. [PMID: 37875116 PMCID: PMC10872926 DOI: 10.1016/j.devcel.2023.10.002] [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: 12/13/2022] [Revised: 08/07/2023] [Accepted: 10/03/2023] [Indexed: 10/26/2023]
Abstract
Mutations in the degradative ubiquitin ligase anaphase-promoting complex (APC) alter neurodevelopment by impairing proteasomal protein clearance, but our understanding of their molecular and cellular pathogenesis remains limited. Here, we employ the proteomic-based discovery of APC substrates in APC mutant mouse brain and human cell lines and identify the chromosome-passenger complex (CPC), topoisomerase 2a (Top2a), and Ki-67 as major chromatin factors targeted by the APC during neuronal differentiation. These substrates accumulate in phosphorylated form, suggesting that they fail to be eliminated after mitosis during terminal differentiation. The accumulation of the CPC kinase Aurora B within constitutive heterochromatin and hyperphosphorylation of its target histone 3 are corrected in the mutant brain by pharmacologic Aurora B inhibition. Surprisingly, the reduction of Ki-67, but not H3S10ph, rescued the function of constitutive heterochromatin in APC mutant neurons. These results expand our understanding of how ubiquitin signaling regulates chromatin during neurodevelopment and identify potential therapeutic targets in APC-related disorders.
Collapse
Affiliation(s)
- Leya Ledvin
- Pathology Department, University of California, San Diego, La Jolla, CA 92093, USA
| | - Brandon M Gassaway
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Jonathan Tawil
- Pathology Department, University of California, San Diego, La Jolla, CA 92093, USA
| | - Olivia Urso
- Pathology Department, University of California, San Diego, La Jolla, CA 92093, USA
| | - Donald Pizzo
- Pathology Department, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kaeli A Welsh
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Derek L Bolhuis
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | | | - Azad Bonni
- Neuroscience Department, Washington University, St. Louis, MO 63110, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Nicholas G Brown
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Cole J Ferguson
- Pathology Department, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
9
|
Titova E, Shagieva G, Dugina V, Kopnin P. The Role of Aurora B Kinase in Normal and Cancer Cells. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:2054-2062. [PMID: 38462449 DOI: 10.1134/s0006297923120088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/22/2023] [Accepted: 10/28/2023] [Indexed: 03/12/2024]
Abstract
Aurora kinases are essential players in mammalian cell division. These kinases are involved in the regulation of spindle dynamics, microtubule-kinetochore interactions, and chromosome condensation and orientation during mitosis. At least three members of the Aurora family - Aurora kinases A, B, and C - have been identified in mammals. Aurora B is essential for maintaining genomic stability and normal cell division. Mutations and dysregulation of this kinase are implicated in tumor initiation and progression. In this review, we discuss the functions of Aurora B, the relationship between increased Aurora B activity and carcinogenesis, and the prospects for the use of Aurora B kinase inhibitors in antitumor therapy.
Collapse
Affiliation(s)
- Ekaterina Titova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| | - Galina Shagieva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Vera Dugina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Pavel Kopnin
- Institute of Carcinogenesis, Blokhin National Medical Research Centre of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| |
Collapse
|
10
|
Peroni E, Randi ML, Rosato A, Cagnin S. Acute myeloid leukemia: from NGS, through scRNA-seq, to CAR-T. dissect cancer heterogeneity and tailor the treatment. J Exp Clin Cancer Res 2023; 42:259. [PMID: 37803464 PMCID: PMC10557350 DOI: 10.1186/s13046-023-02841-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/25/2023] [Indexed: 10/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is a malignant blood cancer with marked cellular heterogeneity due to altered maturation and differentiation of myeloid blasts, the possible causes of which are transcriptional or epigenetic alterations, impaired apoptosis, and excessive cell proliferation. This neoplasm has a high rate of resistance to anticancer therapies and thus a high risk of relapse and mortality because of both the biological diversity of the patient and intratumoral heterogeneity due to the acquisition of new somatic changes. For more than 40 years, the old gold standard "one size fits all" treatment approach included intensive chemotherapy treatment with anthracyclines and cytarabine.The manuscript first traces the evolution of the understanding of the pathology from the 1970s to the present. The enormous strides made in its categorization prove to be crucial for risk stratification, enabling an increasingly personalized diagnosis and treatment approach.Subsequently, we highlight how, over the past 15 years, technological advances enabling single cell RNA sequencing and T-cell modification based on the genomic tools are affecting the classification and treatment of AML. At the dawn of the new millennium, the advent of high-throughput next-generation sequencing technologies has enabled the profiling of patients evidencing different facets of the same disease, stratifying risk, and identifying new possible therapeutic targets that have subsequently been validated. Currently, the possibility of investigating tumor heterogeneity at the single cell level, profiling the tumor at the time of diagnosis or after treatments exist. This would allow the identification of underrepresented cellular subclones or clones resistant to therapeutic approaches and thus responsible for post-treatment relapse that would otherwise be difficult to detect with bulk investigations on the tumor biopsy. Single-cell investigation will then allow even greater personalization of therapy to the genetic and transcriptional profile of the tumor, saving valuable time and dangerous side effects. The era of personalized medicine will take a huge step forward through the disclosure of each individual piece of the complex puzzle that is cancer pathology, to implement a "tailored" therapeutic approach based also on engineered CAR-T cells.
Collapse
Affiliation(s)
- Edoardo Peroni
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, Padova, 35128, Italy.
| | - Maria Luigia Randi
- First Medical Clinic, Department of Medicine-DIMED, University of Padua, Padua, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, Padova, 35128, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Stefano Cagnin
- Department of Biology, University of Padova, Padova, 35131, Italy
- CIR-Myo Myology Center, University of Padova, Padova, 35131, Italy
| |
Collapse
|
11
|
Ren X, Zhuang H, Jiang F, Zhang Y, Zhou P. Barasertib impedes chondrocyte senescence and alleviates osteoarthritis by mitigating the destabilization of heterochromatin induced by AURKB. Biomed Pharmacother 2023; 166:115343. [PMID: 37634474 DOI: 10.1016/j.biopha.2023.115343] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/12/2023] [Accepted: 08/19/2023] [Indexed: 08/29/2023] Open
Abstract
Osteoarthritis (OA) is a common joint disease characterized by progressive cartilage loss that causes disability worldwide. The accumulation of senescent chondrocytes in aging human cartilage contributes to the high incidence of OA. Heterochromatin instability, the hallmark and driving factor of senescence, regulates the expression of the senescence-associated secretory phenotype that induces inflammation and cartilage destruction. However, the role of heterochromatin instability in OA progression remains unclear. In this work, we identified AURKB as a key senescence-associated chromatin regulator using bioinformatics methods. We found that AURKB was upregulated in OA cartilage and chondrocytes exposed to abnormal mechanical strain. Overexpression of AURKB could cause senescence and heterochromatin instability. Furthermore, the AURKB inhibitor Barasertib reversed senescence and heterochromatin instability in chondrocytes and alleviated OA in a rat model. Mechanistically, abnormal mechanical strain increased AURKB levels through the Piezo1/Ca2+ signaling axis. Blocking Piezo1/Ca2+ signaling by short interfering RNA against Piezo1 and Ca2+ chelator BAPTA could reduce the expression of AURKB and alleviate senescence in chondrocytes exposed to abnormal mechanical strain. In conclusion, our data confirmed that abnormal mechanical strain increases the expression of AURKB by activating the Piezo1/Ca2+ signaling axis, leading to destabilized heterochromatin and senescence in chondrocytes, whereas Barasertib consolidates heterochromatin, counteracts senescence and alleviates OA.
Collapse
Affiliation(s)
- Xunshan Ren
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Huangming Zhuang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fuze Jiang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuelong Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Panghu Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
12
|
Kovacs AH, Zhao D, Hou J. Aurora B Inhibitors as Cancer Therapeutics. Molecules 2023; 28:3385. [PMID: 37110619 PMCID: PMC10144992 DOI: 10.3390/molecules28083385] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/29/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
The Aurora kinases (A, B, and C) are a family of three isoform serine/threonine kinases that regulate mitosis and meiosis. The Chromosomal Passenger Complex (CPC), which contains Aurora B as an enzymatic component, plays a critical role in cell division. Aurora B in the CPC ensures faithful chromosome segregation and promotes the correct biorientation of chromosomes on the mitotic spindle. Aurora B overexpression has been observed in several human cancers and has been associated with a poor prognosis for cancer patients. Targeting Aurora B with inhibitors is a promising therapeutic strategy for cancer treatment. In the past decade, Aurora B inhibitors have been extensively pursued in both academia and industry. This paper presents a comprehensive review of the preclinical and clinical candidates of Aurora B inhibitors as potential anticancer drugs. The recent advances in the field of Aurora B inhibitor development will be highlighted, and the binding interactions between Aurora B and inhibitors based on crystal structures will be presented and discussed to provide insights for the future design of more selective Aurora B inhibitors.
Collapse
Affiliation(s)
- Antal H. Kovacs
- Department of Chemistry, Lakehead University, 955 Oliver Road, Thunder Bay, ON P7B 5E1, Canada
| | - Dong Zhao
- Department of Chemistry, Lakehead University, 955 Oliver Road, Thunder Bay, ON P7B 5E1, Canada
| | - Jinqiang Hou
- Department of Chemistry, Lakehead University, 955 Oliver Road, Thunder Bay, ON P7B 5E1, Canada
- Thunder Bay Regional Health Research Institute, 980 Oliver Road, Thunder Bay, ON P7B 6V4, Canada
| |
Collapse
|
13
|
Aurora B Kinase Inhibition by AZD1152 Concomitant with Tumor Treating Fields Is Effective in the Treatment of Cultures from Primary and Recurrent Glioblastomas. Int J Mol Sci 2023; 24:ijms24055016. [PMID: 36902447 PMCID: PMC10003311 DOI: 10.3390/ijms24055016] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Tumor Treating Fields (TTFields) were incorporated into the treatment of glioblastoma, the most malignant brain tumor, after showing an effect on progression-free and overall survival in a phase III clinical trial. The combination of TTFields and an antimitotic drug might further improve this approach. Here, we tested the combination of TTFields with AZD1152, an Aurora B kinase inhibitor, in primary cultures of newly diagnosed (ndGBM) and recurrent glioblastoma (rGBM). AZD1152 concentration was titrated for each cell line and 5-30 nM were used alone or in addition to TTFields (1.6 V/cm RMS; 200 kHz) applied for 72 h using the inovitro™ system. Cell morphological changes were visualized by conventional and confocal laser microscopy. The cytotoxic effects were determined by cell viability assays. Primary cultures of ndGBM and rGBM varied in p53 mutational status; ploidy; EGFR expression and MGMT-promoter methylation status. Nevertheless; in all primary cultures; a significant cytotoxic effect was found following TTFields treatment alone and in all but one, a significant effect after treatment with AZD1152 alone was also observed. Moreover, in all primary cultures the combined treatment had the most pronounced cytotoxic effect in parallel with morphological changes. The combined treatment of TTFields and AZD1152 led to a significant reduction in the number of ndGBM and rGBM cells compared to each treatment alone. Further evaluation of this approach, which has to be considered as a proof of concept, is warranted, before entering into early clinical trials.
Collapse
|
14
|
Johnson ML, Wang JS, Falchook G, Greenlees C, Jones S, Strickland D, Fabbri G, Kennedy C, Elizabeth Pease J, Sainsbury L, MacDonald A, Schalkwijk S, Szekeres P, Cosaert J, Burris H. Safety, tolerability, and pharmacokinetics of Aurora kinase B inhibitor AZD2811: a phase 1 dose-finding study in patients with advanced solid tumours. Br J Cancer 2023; 128:1906-1915. [PMID: 36871042 PMCID: PMC10147685 DOI: 10.1038/s41416-023-02185-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 01/18/2023] [Accepted: 01/25/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND AZD2811 is a potent, selective Aurora kinase B inhibitor. We report the dose-escalation phase of a first-in-human study assessing nanoparticle-encapsulated AZD2811 in advanced solid tumours. METHODS AZD2811 was administered in 12 dose-escalation cohorts (2-h intravenous infusion; 15‒600 mg; 21-/28-day cycles) with granulocyte colony-stimulating factor (G-CSF) at higher doses. The primary objective was determining safety and maximum tolerated/recommended phase 2 dose (RP2D). RESULTS Fifty-one patients received AZD2811. Drug exposure was sustained for several days post-dose. The most common AZD2811-related adverse events (AEs) were fatigue (27.3%) at ≤200 mg/cycle and neutropenia (37.9%) at ≥400 mg/cycle. Five patients had dose-limiting toxicities: grade (G)4 decreased neutrophil count (n = 1, 200 mg; Days 1, 4; 28-day cycle); G4 decreased neutrophil count and G3 stomatitis (n = 1 each, both 400 mg; Day 1; 21-day cycle); G3 febrile neutropenia and G3 fatigue (n = 1 each, both 600 mg; Day 1; 21-day cycle +G-CSF). RP2D was 500 mg; Day 1; 21-day cycle with G-CSF on Day 8. Neutropenia/neutrophil count decrease were on-target AEs. Best overall responses were partial response (n = 1, 2.0%) and stable disease (n = 23, 45.1%). CONCLUSIONS At RP2D, AZD2811 was tolerable with G-CSF support. Neutropenia was a pharmacodynamic biomarker. CLINICAL TRIAL REGISTRATION NCT02579226.
Collapse
Affiliation(s)
- Melissa L Johnson
- Sarah Cannon Research Institute, Nashville, TN, USA. .,Tennessee Oncology, Nashville, TN, USA.
| | - Judy S Wang
- Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota, FL, USA
| | - Gerald Falchook
- Sarah Cannon Research Institute at HealthONE, Denver, CO, USA
| | - Carol Greenlees
- Sarah Cannon Research Institute, Nashville, TN, USA.,Avacta Life Sciences, London, UK
| | | | | | | | | | | | | | | | | | | | | | - Howard Burris
- Sarah Cannon Research Institute, Nashville, TN, USA.,Tennessee Oncology, Nashville, TN, USA
| |
Collapse
|
15
|
Stefani A, Piro G, Schietroma F, Strusi A, Vita E, Fiorani S, Barone D, Monaca F, Sparagna I, Valente G, Ferrara MG, D’Argento E, Di Salvatore M, Carbone C, Tortora G, Bria E. Unweaving the mitotic spindle: A focus on Aurora kinase inhibitors in lung cancer. Front Oncol 2022; 12:1026020. [PMID: 36387232 PMCID: PMC9647054 DOI: 10.3389/fonc.2022.1026020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/17/2022] [Indexed: 07/30/2023] Open
Abstract
Lung cancer is one of the most aggressive malignancies, classified into two major histological subtypes: non-small cell lung cancer (NSCLC), that accounts for about 85% of new diagnosis, and small cell lung cancer (SCLC), the other 15%. In the case of NSCLC, comprehensive genome sequencing has allowed the identification of an increasing number of actionable targets, which have become the cornerstone of treatment in the advanced setting. On the other hand, the concept of oncogene-addiction is lacking in SCLC, and the only innovation of the last 30 years has been the introduction of immune checkpoint inhibitors in extensive stage disease. Dysregulation of cell cycle is a fundamental step in carcinogenesis, and Aurora kinases (AURKs) are a family of serine/threonine kinases that play a crucial role in the correct advance through the steps of the cycle. Hyperexpression of Aurora kinases is a common protumorigenic pathway in many cancer types, including NSCLC and SCLC; in addition, different mechanisms of resistance to anticancer drugs rely on AURK expression. Hence, small molecule inhibitors of AURKs have been developed in recent years and tested in several malignancies, with different results. The aim of this review is to analyze the current evidences of AURK inhibition in lung cancer, starting from preclinical rationale to finish with clinical trials available up to now.
Collapse
Affiliation(s)
- Alessio Stefani
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Geny Piro
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesco Schietroma
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Strusi
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Emanuele Vita
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Simone Fiorani
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Diletta Barone
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Federico Monaca
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ileana Sparagna
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giustina Valente
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Miriam Grazia Ferrara
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ettore D’Argento
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Mariantonietta Di Salvatore
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carmine Carbone
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giampaolo Tortora
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Emilio Bria
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Section of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
16
|
Komatsu M, Nakamura K, Takeda T, Chiwaki F, Banno K, Aoki D, Takeshita F, Sasaki H. Aurora kinase blockade drives de novo addiction of cervical squamous cell carcinoma to druggable EGFR signalling. Oncogene 2022; 41:2326-2339. [DOI: 10.1038/s41388-022-02256-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/08/2022] [Accepted: 02/17/2022] [Indexed: 12/24/2022]
|
17
|
Lewuillon C, Laguillaumie MO, Quesnel B, Idziorek T, Touil Y, Lemonnier L. Put in a “Ca2+ll” to Acute Myeloid Leukemia. Cells 2022; 11:cells11030543. [PMID: 35159351 PMCID: PMC8834247 DOI: 10.3390/cells11030543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/28/2022] [Accepted: 02/01/2022] [Indexed: 02/05/2023] Open
Abstract
Acute myeloid leukemia (AML) is a clonal disorder characterized by genetic aberrations in myeloid primitive cells (blasts) which lead to their defective maturation/function and their proliferation in the bone marrow (BM) and blood of affected individuals. Current intensive chemotherapy protocols result in complete remission in 50% to 80% of AML patients depending on their age and the AML type involved. While alterations in calcium signaling have been extensively studied in solid tumors, little is known about the role of calcium in most hematologic malignancies, including AML. Our purpose with this review is to raise awareness about this issue and to present (i) the role of calcium signaling in AML cell proliferation and differentiation and in the quiescence of hematopoietic stem cells; (ii) the interplay between mitochondria, metabolism, and oxidative stress; (iii) the effect of the BM microenvironment on AML cell fate; and finally (iv) the mechanism by which chemotherapeutic treatments modify calcium homeostasis in AML cells.
Collapse
Affiliation(s)
- Clara Lewuillon
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Marie-Océane Laguillaumie
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Bruno Quesnel
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Thierry Idziorek
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Yasmine Touil
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Loïc Lemonnier
- Univ. Lille, Inserm, U1003—PHYCEL—Physiologie Cellulaire, F-59000 Lille, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, F-59655 Villeneuve d’Ascq, France
- Correspondence:
| |
Collapse
|
18
|
Llombart V, Mansour MR. Therapeutic targeting of "undruggable" MYC. EBioMedicine 2022; 75:103756. [PMID: 34942444 PMCID: PMC8713111 DOI: 10.1016/j.ebiom.2021.103756] [Citation(s) in RCA: 257] [Impact Index Per Article: 85.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/23/2021] [Accepted: 12/01/2021] [Indexed: 12/13/2022] Open
Abstract
c-MYC controls global gene expression and regulates cell proliferation, cell differentiation, cell cycle, metabolism and apoptosis. According to some estimates, MYC is dysregulated in ≈70% of human cancers and strong evidence implicates aberrantly expressed MYC in both tumor initiation and maintenance. In vivo studies show that MYC inhibition elicits a prominent anti-proliferative effect and sustained tumor regression while any alteration on healthy tissue remains reversible. This opens an exploitable window for treatment that makes MYC one of the most appealing therapeutic targets for cancer drug development. This review describes the main functional and structural features of the protein structure of MYC and provides a general overview of the most relevant or recently identified interactors that modulate MYC oncogenic activity. This review also summarizes the different approaches aiming to abrogate MYC oncogenic function, with a particular focus on the prototype inhibitors designed for the direct and indirect targeting of MYC.
Collapse
Affiliation(s)
- Victor Llombart
- UCL Cancer Institute, University College London, Department of Haematology, London WC1E 6DD, UK
| | - Marc R Mansour
- UCL Cancer Institute, University College London, Department of Haematology, London WC1E 6DD, UK; UCL Great Ormond Street Institute of Child Health, Developmental Biology and Cancer, London, UK.
| |
Collapse
|
19
|
Tariq MU, Furqan M, Parveen H, Ullah R, Muddassar M, Saleem RSZ, Bavetsias V, Linardopoulos S, Faisal A. CCT245718, a dual FLT3/Aurora A inhibitor overcomes D835Y-mediated resistance to FLT3 inhibitors in acute myeloid leukaemia cells. Br J Cancer 2021; 125:966-974. [PMID: 34446858 DOI: 10.1038/s41416-021-01527-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/17/2021] [Accepted: 08/11/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Activating mutations in the Fms-like tyrosine kinase 3 (FLT3) are among the most prevalent oncogenic mutations in acute myeloid leukaemia. Inhibitors selectively targeting FLT3 kinase have shown promising clinical activity; their success in the clinic, however, has been limited due to the emergence of acquired resistance. METHODS CCT245718 was identified and characterised as a dual Aurora A/FLT3 inhibitor through cell-based and biochemical assays. The ability of CCT245718 to overcome TKD-mediated resistance was evaluated in a cell line-based model of drug resistance to FLT3 inhibitors. RESULTS CCT245718 exhibits potent antiproliferative activity towards FLT3-ITD + AML cell lines and strongly binds to FLT3-ITD and TKD (D835Y) mutants in vitro. Activities of both FLT3-ITD and Aurora A are also inhibited in cells. Inhibition of FLT3 results in reduced phosphorylation of STAT5, downregulation of survivin and induction of apoptotic cell death. Moreover, CCT245718 overcomes TKD-mediated resistance in a MOLM-13-derived cell line containing FLT3 with both ITD and D835Y mutations. It also inhibits FLT3 signalling in both parental and resistant cell lines compared to FLT3-specific inhibitor MLN518, which is only active in the parental cell line. CONCLUSIONS Our results demonstrate that CCT245718 is a potent dual FLT3/Aurora A inhibitor that can overcome TKD-mediated acquired resistance.
Collapse
Affiliation(s)
- Muhammad Usama Tariq
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Muhammad Furqan
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Hira Parveen
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Rahim Ullah
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Muhammad Muddassar
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Rahman Shah Zaib Saleem
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Vassilios Bavetsias
- Cancer Research UK, Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Spiros Linardopoulos
- Cancer Research UK, Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK.,Breast Cancer Now, Division of Breast Cancer Research, The Institute of Cancer Research, London, UK.,AstraZeneca, Cambridge, UK
| | - Amir Faisal
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan.
| |
Collapse
|
20
|
Novais P, Silva PMA, Amorim I, Bousbaa H. Second-Generation Antimitotics in Cancer Clinical Trials. Pharmaceutics 2021; 13:1011. [PMID: 34371703 PMCID: PMC8309102 DOI: 10.3390/pharmaceutics13071011] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/24/2021] [Accepted: 06/29/2021] [Indexed: 12/17/2022] Open
Abstract
Mitosis represents a promising target to block cancer cell proliferation. Classical antimitotics, mainly microtubule-targeting agents (MTAs), such as taxanes and vinca alkaloids, are amongst the most successful anticancer drugs. By disrupting microtubules, they activate the spindle assembly checkpoint (SAC), which induces a prolonged delay in mitosis, expected to induce cell death. However, resistance, toxicity, and slippage limit the MTA's effectiveness. With the desire to overcome some of the MTA's limitations, mitotic and SAC components have attracted great interest as promising microtubule-independent targets, leading to the so-called second-generation antimitotics (SGAs). The identification of inhibitors against most of these targets, and the promising outcomes achieved in preclinical assays, has sparked the interest of academia and industry. Many of these inhibitors have entered clinical trials; however, they exhibited limited efficacy as monotherapy, and failed to go beyond phase II trials. Combination therapies are emerging as promising strategies to give a second chance to these SGAs. Here, an updated view of the SGAs that reached clinical trials is here provided, together with future research directions, focusing on inhibitors that target the SAC components.
Collapse
Affiliation(s)
- Pedro Novais
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (P.N.); (P.M.A.S.)
- Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Patrícia M. A. Silva
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (P.N.); (P.M.A.S.)
| | - Isabel Amorim
- GreenUPorto (Sustainable Agrifood Production) Research Center, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal;
| | - Hassan Bousbaa
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (P.N.); (P.M.A.S.)
| |
Collapse
|
21
|
Guo C, Gao YY, Ju QQ, Zhang CX, Gong M, Li ZL. The landscape of gene co-expression modules correlating with prognostic genetic abnormalities in AML. J Transl Med 2021; 19:228. [PMID: 34051812 PMCID: PMC8164775 DOI: 10.1186/s12967-021-02914-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022] Open
Abstract
Background The heterogenous cytogenetic and molecular variations were harbored by AML patients, some of which are related with AML pathogenesis and clinical outcomes. We aimed to uncover the intrinsic expression profiles correlating with prognostic genetic abnormalities by WGCNA. Methods We downloaded the clinical and expression dataset from BeatAML, TCGA and GEO database. Using R (version 4.0.2) and ‘WGCNA’ package, the co-expression modules correlating with the ELN2017 prognostic markers were identified (R2 ≥ 0.4, p < 0.01). ORA detected the enriched pathways for the key co-expression modules. The patients in TCGA cohort were randomly assigned into the training set (50%) and testing set (50%). The LASSO penalized regression analysis was employed to build the prediction model, fitting OS to the expression level of hub genes by ‘glmnet’ package. Then the testing and 2 independent validation sets (GSE12417 and GSE37642) were used to validate the diagnostic utility and accuracy of the model. Results A total of 37 gene co-expression modules and 973 hub genes were identified for the BeatAML cohort. We found that 3 modules were significantly correlated with genetic markers (the ‘lightyellow’ module for NPM1 mutation, the ‘saddlebrown’ module for RUNX1 mutation, the ‘lightgreen’ module for TP53 mutation). ORA revealed that the ‘lightyellow’ module was mainly enriched in DNA-binding transcription factor activity and activation of HOX genes. The ‘saddlebrown’ module was enriched in immune response process. And the ‘lightgreen’ module was predominantly enriched in mitosis cell cycle process. The LASSO- regression analysis identified 6 genes (NFKB2, NEK9, HOXA7, APRC5L, FAM30A and LOC105371592) with non-zero coefficients. The risk score generated from the 6-gene model, was associated with ELN2017 risk stratification, relapsed disease, and prior MDS history. The 5-year AUC for the model was 0.822 and 0.824 in the training and testing sets, respectively. Moreover, the diagnostic utility of the model was robust when it was employed in 2 validation sets (5-year AUC 0.743–0.79). Conclusions We established the co-expression network signature correlated with the ELN2017 recommended prognostic genetic abnormalities in AML. The 6-gene prediction model for AML survival was developed and validated by multiple datasets. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02914-2.
Collapse
Affiliation(s)
- Chao Guo
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, China
| | - Ya-Yue Gao
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, China
| | - Qian-Qian Ju
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, China
| | - Chun-Xia Zhang
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, China
| | - Ming Gong
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, China
| | - Zhen-Ling Li
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, China.
| |
Collapse
|
22
|
de Oliveira Lisboa M, Brofman PRS, Schmid-Braz AT, Rangel-Pozzo A, Mai S. Chromosomal Instability in Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:cancers13112655. [PMID: 34071283 PMCID: PMC8198625 DOI: 10.3390/cancers13112655] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 12/20/2022] Open
Abstract
Chromosomal instability (CIN), the increasing rate in which cells acquire new chromosomal alterations, is one of the hallmarks of cancer. Many studies highlighted CIN as an important mechanism in the origin, progression, and relapse of acute myeloid leukemia (AML). The ambivalent feature of CIN as a cancer-promoting or cancer-suppressing mechanism might explain the prognostic variability. The latter, however, is described in very few studies. This review highlights the important CIN mechanisms in AML, showing that CIN signatures can occur largely in all the three major AML types (de novo AML, secondary-AML, and therapy-related-AML). CIN features in AML could also be age-related and reflect the heterogeneity of the disease. Although most of these abnormalities show an adverse prognostic value, they also offer a strong new perspective on personalized therapy approaches, which goes beyond assessing CIN in vitro in patient tumor samples to predict prognosis. Current and emerging AML therapies are exploring CIN to improve AML treatment, which includes blocking CIN or increasing CIN beyond the limit threshold to induce cell death. We argue that the characterization of CIN features, not included yet in the routine diagnostic of AML patients, might provide a better stratification of patients and be extended to a more personalized therapeutic approach.
Collapse
Affiliation(s)
- Mateus de Oliveira Lisboa
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná—PUCPR, Curitiba 80215-901, Paraná, Brazil; (M.d.O.L.); (P.R.S.B.)
| | - Paulo Roberto Slud Brofman
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná—PUCPR, Curitiba 80215-901, Paraná, Brazil; (M.d.O.L.); (P.R.S.B.)
| | - Ana Teresa Schmid-Braz
- Hospital das Clínicas, Universidade Federal do Paraná, Curitiba 80060-240, Paraná, Brazil;
| | - Aline Rangel-Pozzo
- Department of Physiology and Pathophysiology, University of Manitoba, Cell Biology, CancerCare Manitoba Research Institute, Winnipeg, MB R3C 2B7, Canada
- Correspondence: (A.R.-P.); (S.M.); Tel.: +1-(204)787-4125 (S.M.)
| | - Sabine Mai
- Department of Physiology and Pathophysiology, University of Manitoba, Cell Biology, CancerCare Manitoba Research Institute, Winnipeg, MB R3C 2B7, Canada
- Correspondence: (A.R.-P.); (S.M.); Tel.: +1-(204)787-4125 (S.M.)
| |
Collapse
|
23
|
Aurora Kinase B Inhibition: A Potential Therapeutic Strategy for Cancer. Molecules 2021; 26:molecules26071981. [PMID: 33915740 PMCID: PMC8037052 DOI: 10.3390/molecules26071981] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/23/2022] Open
Abstract
Aurora kinase B (AURKB) is a mitotic serine/threonine protein kinase that belongs to the aurora kinase family along with aurora kinase A (AURKA) and aurora kinase C (AURKC). AURKB is a member of the chromosomal passenger protein complex and plays a role in cell cycle progression. Deregulation of AURKB is observed in several tumors and its overexpression is frequently linked to tumor cell invasion, metastasis and drug resistance. AURKB has emerged as an attractive drug target leading to the development of small molecule inhibitors. This review summarizes recent findings pertaining to the role of AURKB in tumor development, therapy related drug resistance, and its inhibition as a potential therapeutic strategy for cancer. We discuss AURKB inhibitors that are in preclinical and clinical development and combination studies of AURKB inhibition with other therapeutic strategies.
Collapse
|
24
|
Wang C, Zhang J, Yin J, Gan Y, Xu S, Gu Y, Huang W. Alternative approaches to target Myc for cancer treatment. Signal Transduct Target Ther 2021; 6:117. [PMID: 33692331 PMCID: PMC7946937 DOI: 10.1038/s41392-021-00500-y] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 12/07/2020] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
The Myc proto-oncogene family consists of three members, C-MYC, MYCN, and MYCL, which encodes the transcription factor c-Myc (hereafter Myc), N-Myc, and L-Myc, respectively. Myc protein orchestrates diverse physiological processes, including cell proliferation, differentiation, survival, and apoptosis. Myc modulates about 15% of the global transcriptome, and its deregulation rewires the cellular signaling modules inside tumor cells, thereby acquiring selective advantages. The deregulation of Myc occurs in >70% of human cancers, and is related to poor prognosis; hence, hyperactivated Myc oncoprotein has been proposed as an ideal drug target for decades. Nevertheless, no specific drug is currently available to directly target Myc, mainly because of its "undruggable" properties: lack of enzymatic pocket for conventional small molecules to bind; inaccessibility for antibody due to the predominant nucleus localization of Myc. Although the topic of targeting Myc has actively been reviewed in the past decades, exciting new progresses in this field keep emerging. In this review, after a comprehensive summarization of valuable sources for potential druggable targets of Myc-driven cancer, we also peer into the promising future of utilizing macropinocytosis to deliver peptides like Omomyc or antibody agents to intracellular compartment for cancer treatment.
Collapse
Affiliation(s)
- Chen Wang
- Division of Medical Genomics and Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Genetics, Zhejiang University and Department of Genetics, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Lab of Genetic and Developmental Disorder, Hangzhou, Zhejiang, 310058, China
| | - Jiawei Zhang
- Division of Medical Genomics and Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jie Yin
- Division of Medical Genomics and Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Genetics, Zhejiang University and Department of Genetics, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Lab of Genetic and Developmental Disorder, Hangzhou, Zhejiang, 310058, China
| | - Yichao Gan
- Division of Medical Genomics and Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Genetics, Zhejiang University and Department of Genetics, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Lab of Genetic and Developmental Disorder, Hangzhou, Zhejiang, 310058, China
| | - Senlin Xu
- Molecular and Cellular Biology of Cancer Program & Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, CA, USA
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Ying Gu
- Division of Medical Genomics and Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Institute of Genetics, Zhejiang University and Department of Genetics, School of Medicine, Zhejiang University, Hangzhou, China.
- Zhejiang Provincial Key Lab of Genetic and Developmental Disorder, Hangzhou, Zhejiang, 310058, China.
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 311121, China.
| | - Wendong Huang
- Molecular and Cellular Biology of Cancer Program & Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, CA, USA.
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
25
|
Lin X, Xiang X, Hao L, Wang T, Lai Y, Abudoureyimu M, Zhou H, Feng B, Chu X, Wang R. The role of Aurora-A in human cancers and future therapeutics. Am J Cancer Res 2020; 10:2705-2729. [PMID: 33042612 PMCID: PMC7539775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/05/2020] [Indexed: 06/11/2023] Open
Abstract
Aurora-A is a mitotic serine/threonine-protein kinase and an oncogene. In normal cells, Aurora-A appears from G2 phase and localizes at the centrosome, where it participates in centrosome replication, isolation and maturation. Aurora-A also maintains Golgi apparatus structure and spindle assembly. Aurora-A undergoes ubiquitination-mediated degradation after the cell division phase. Aurora-A is abnormally expressed in tumor cells and promotes cell proliferation by regulating mitotic substrates, such as PP1, PLK1, TPX2, and LAST2, and affects other molecules through a non-mitotic pathway to promote cell invasion and metastasis. Some molecules in tumor cells also indirectly act on Aurora-A to regulate tumor cells. Aurora-A also mediates resistance to chemotherapy and radiotherapy and is involved in tumor immunotherapy. Clinical trials of Aurora-A molecular inhibitors are currently underway, and clinical transformation is just around the corner.
Collapse
Affiliation(s)
- Xinrong Lin
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing UniversityNanjing, China
| | - Xiaosong Xiang
- Affiliated Jinling Hospital Research Institution of General Surgery, Medical School of Nanjing UniversityNanjing, China
| | - Liping Hao
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing UniversityNanjing, China
| | - Ting Wang
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing UniversityNanjing, China
| | - Yongting Lai
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, First School of Clinical Medicine, Southern Medical UniversityNanjing, China
| | - Mubalake Abudoureyimu
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing UniversityNanjing, China
| | - Hao Zhou
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing UniversityNanjing, China
| | - Bing Feng
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing UniversityNanjing, China
| | - Xiaoyuan Chu
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing UniversityNanjing, China
| | - Rui Wang
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing UniversityNanjing, China
| |
Collapse
|
26
|
Kasam RK, Ghandikota S, Soundararajan D, Reddy GB, Huang SK, Jegga AG, Madala SK. Inhibition of Aurora Kinase B attenuates fibroblast activation and pulmonary fibrosis. EMBO Mol Med 2020; 12:e12131. [PMID: 32761869 PMCID: PMC7507328 DOI: 10.15252/emmm.202012131] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
Fibroblast activation including proliferation, survival, and ECM production is central to initiation and maintenance of fibrotic lesions in idiopathic pulmonary fibrosis (IPF). However, druggable molecules that target fibroblast activation remain limited. In this study, we show that multiple pro‐fibrotic growth factors, including TGFα, CTGF, and IGF1, increase aurora kinase B (AURKB) expression and activity in fibroblasts. Mechanistically, we demonstrate that Wilms tumor 1 (WT1) is a key transcription factor that mediates TGFα‐driven AURKB upregulation in fibroblasts. Importantly, we found that inhibition of AURKB expression or activity is sufficient to attenuate fibroblast activation. We show that fibrosis induced by TGFα is highly dependent on AURKB expression and treating TGFα mice with barasertib, an AURKB inhibitor, reverses fibroblast activation, and pulmonary fibrosis. Barasertib similarly attenuated fibrosis in the bleomycin model of pulmonary fibrosis. Together, our preclinical studies provide important proof‐of‐concept that demonstrate barasertib as a possible intervention therapy for IPF.
Collapse
Affiliation(s)
- Rajesh K Kasam
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Biochemistry, National Institute of Nutrition, Hyderabad, India
| | - Sudhir Ghandikota
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Computer Science, University of Cincinnati College of Engineering, Cincinnati, OH, USA
| | | | - Geereddy B Reddy
- Department of Biochemistry, National Institute of Nutrition, Hyderabad, India
| | - Steven K Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anil G Jegga
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Computer Science, University of Cincinnati College of Engineering, Cincinnati, OH, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Satish K Madala
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
27
|
Liu M, Ju X, Zou J, Shi J, Jia G. Recent researches for dual Aurora target inhibitors in antitumor field. Eur J Med Chem 2020; 203:112498. [PMID: 32693295 DOI: 10.1016/j.ejmech.2020.112498] [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: 03/21/2020] [Revised: 05/05/2020] [Accepted: 05/28/2020] [Indexed: 11/17/2022]
Abstract
Non-infectious and chronic diseases such as malignant tumors are now one of the main causes of human death. Its occurrence is a multi-factor, multi-step complex process with biological characteristics such as cell differentiation, abnormal proliferation, uncontrolled growth, and metastasis. It has been found that a variety of human malignant tumors are accompanied by over-expression and proliferation of Aurora kinase, which causes abnormalities in the mitotic process and is related to the instability of the genome that causes tumors. Therefore, the use of Aurora kinase inhibitors to target tumors is becoming a research hotspot. However, in cancer, because of the complexity of signal transduction system and the participation of different proteins and enzymes, the anticancer effect of selective single-target drugs is limited. After inhibiting one pathway, signal molecules can be conducted through other pathways, resulting in poor therapeutic effect of single-target drug treatment. Multi-target drugs can solve this problem very well. It can regulate the various links that cause disease at the same time without completely eliminating the relationship between the signal transmission systems, and it is not easy to cause drug resistance. Currently, studies have shown that Aurora dual-target inhibitors generated with the co-inhibition of Aurora and another target (such as CDK, PLK, JAK2, etc.) have better therapeutic effects on tumors. In this paper, we reviewed the studies of dual Aurora inhibitors that have been discovered in recent years.
Collapse
Affiliation(s)
- Maoyu Liu
- The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicines of Ministry, State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xueming Ju
- Department of Ultrasound, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jing Zou
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Guiqing Jia
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
28
|
Green SD, Konig H. Treatment of Acute Myeloid Leukemia in the Era of Genomics-Achievements and Persisting Challenges. Front Genet 2020; 11:480. [PMID: 32536937 PMCID: PMC7267060 DOI: 10.3389/fgene.2020.00480] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/17/2020] [Indexed: 01/15/2023] Open
Abstract
Acute myeloid leukemia (AML) represents a malignant disorder of the hematopoietic system that is mainly characterized by rapid proliferation, dysregulated apoptosis, and impaired differentiation of leukemic blasts. For several decades, the diagnostic approach in AML was largely based on histologic characteristics with little impact on the treatment decision-making process. This perspective has drastically changed within the past years due to the advent of novel molecular technologies, such as whole genome next-generation sequencing (NGS), and the resulting knowledge gain in AML biology and pathogenesis. After more than four decades of intensive chemotherapy as a "one-size-fits-all" concept, several targeted agents have recently been approved for the treatment of AML, either as single agents or as part of combined treatment regimens. Several other compounds, directed against regulators of apoptotic, epigenetic, or microenvironmental pathways, as well as modulators of the immune system, are currently in development and being investigated in clinical trials. The constant progress in AML research has started to produce improved survival rates and fueled hopes that a once rapidly fatal disease can be transformed into a chronic condition. In this review, the authors provide a summary of recent advances in the development of targeted AML therapies and discuss persistent challenges.
Collapse
Affiliation(s)
| | - Heiko Konig
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, United States
| |
Collapse
|
29
|
Wang JX, Zhang L, Huang ZW, Zhang XN, Jiang YY, Liu FJ, Long L, Xue MJ, Lu G, Liu Q, Long ZJ. Aurora kinase inhibitor restrains STAT5-activated leukemic cell proliferation by inducing mitochondrial impairment. J Cell Physiol 2020; 235:8358-8370. [PMID: 32239704 DOI: 10.1002/jcp.29680] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/13/2020] [Indexed: 12/21/2022]
Abstract
Current chemotherapy regimens on acute myeloid leukemia (AML) still have some drawbacks, such as intolerance and drug resistance, which calls need for the development of targeted therapy. Signal transducer and activator of transcription 5 (STAT5) is often overexpressed or abnormally activated in leukemia and involved in cell self-renewal, proliferation, and stress adaptation. Overexpressed Aurora A (AURKA) is associated with poor prognosis in tumors, and inhibitors against AURKA are already in clinical trials. However, it has rarely been reported whether AURKA inhibitors restrain STAT5-activated leukemia cells. In this study, we constructed STAT5 constitutively activated (cS5) cells and found that STAT5 promoted cell proliferation and colony formation. Moreover, cS5 cells showed elevated reactive oxygen species (ROS) and adenosine triphosphate (ATP) levels, which indicated higher mitochondrial metabolism in cS5 cells. A novel AURKA inhibitor AKI604 was synthesized and showed significant inhibitory effects to the proliferation and colony formation in both STAT5 constitutively activated and nonactivated AML cells. AKI604 induced mitochondrial impairment, leading to the disruption of mitochondrial membrane potential and the elevation of ROS as well as cellular calcium (Ca2+ ) levels. AKI604 could also decline basal oxygen consumption rate and ATP biosynthesis, indicating the damage of oxidative phosphorylation. Furthermore, AKI604 exhibited significant antitumor effect in the HL-60 cS5 xenograft model of the BALB/c nude mice without an obvious influence on mice body weight and other healthy indicators. This study suggested that AKI604 was a potential strategy to overcome STAT5-induced leukemic proliferation in AML treatment by inducing mitochondrial impairment.
Collapse
Affiliation(s)
- Jin-Xing Wang
- Department of Hematology, The Third Affiliated Hospital, Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Ling Zhang
- Department of Hematology, The Third Affiliated Hospital, Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Ze-Wei Huang
- Department of Hematology, The Third Affiliated Hospital, Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Xue-Ning Zhang
- Department of Hematology, The Third Affiliated Hospital, Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Yan-Yan Jiang
- Department of Hematology, The Third Affiliated Hospital, Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Fang-Jie Liu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Liang Long
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Man-Jie Xue
- Medical Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Gui Lu
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Quentin Liu
- Department of Hematology, The Third Affiliated Hospital, Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Zi-Jie Long
- Department of Hematology, The Third Affiliated Hospital, Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
30
|
Jiang J, Wang J, Yue M, Cai X, Wang T, Wu C, Su H, Wang Y, Han M, Zhang Y, Zhu X, Jiang P, Li P, Sun Y, Xiao W, Feng H, Qing G, Liu H. Direct Phosphorylation and Stabilization of MYC by Aurora B Kinase Promote T-cell Leukemogenesis. Cancer Cell 2020; 37:200-215.e5. [PMID: 32049046 PMCID: PMC7321798 DOI: 10.1016/j.ccell.2020.01.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 10/15/2019] [Accepted: 01/07/2020] [Indexed: 12/12/2022]
Abstract
Deregulation of MYC plays an essential role in T cell acute lymphoblastic leukemia (T-ALL), yet the mechanisms underlying its deregulation remain elusive. Herein, we identify a molecular mechanism responsible for reciprocal activation between Aurora B kinase (AURKB) and MYC. AURKB directly phosphorylates MYC at serine 67, counteracting GSK3β-directed threonine 58 phosphorylation and subsequent FBXW7-mediated proteasomal degradation. Stabilized MYC, in concert with T cell acute lymphoblastic leukemia 1 (TAL1), directly activates AURKB transcription, constituting a positive feedforward loop that reinforces MYC-regulated oncogenic programs. Therefore, inhibitors of AURKB induce prominent MYC degradation concomitant with robust leukemia cell death. These findings reveal an AURKB-MYC regulatory circuit that underlies T cell leukemogenesis, and provide a rationale for therapeutic targeting of oncogenic MYC via AURKB inhibition.
Collapse
Affiliation(s)
- Jue Jiang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Jingchao Wang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Ming Yue
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Xiaolian Cai
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan 430072, China
| | - Tianci Wang
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Chao Wu
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Hexiu Su
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Yanwu Wang
- Department of Histology and Embryology, School of Basic Medical Science, Wuhan University, Wuhan 430071, China
| | - Meng Han
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yingchi Zhang
- State Key Laboratory of Experimental Hematology and Division of Pediatric Blood Diseases Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Xiaofan Zhu
- State Key Laboratory of Experimental Hematology and Division of Pediatric Blood Diseases Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Peng Jiang
- School of Life Sciences, Tsinghua University, Collaborative Innovation Center for Cancer Medicine, Beijing 100084, China
| | - Peng Li
- South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yonghua Sun
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, Cancer Research Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Wuhan Xiao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan 430072, China
| | - Hui Feng
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, Cancer Research Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Guoliang Qing
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Hudan Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
31
|
Manzione MG, Rombouts J, Steklov M, Pasquali L, Sablina A, Gelens L, Qian J, Bollen M. Co-regulation of the antagonistic RepoMan:Aurora-B pair in proliferating cells. Mol Biol Cell 2020; 31:419-438. [PMID: 31967936 PMCID: PMC7185888 DOI: 10.1091/mbc.e19-12-0698] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chromosome segregation during mitosis is antagonistically regulated by the Aurora-B kinase and RepoMan (recruits PP1 onto mitotic chromatin at anaphase)-associated phosphatases PP1/PP2A. Aurora B is overexpressed in many cancers but, surprisingly, this only rarely causes lethal aneuploidy. Here we show that RepoMan abundance is regulated by the same mechanisms that control Aurora B, including FOXM1-regulated expression and proteasomal degradation following ubiquitination by APC/C-CDH1 or SCFFBXW7. The deregulation of these mechanisms can account for the balanced co-overexpression of Aurora B and RepoMan in many cancers, which limits chromosome segregation errors. In addition, Aurora B and RepoMan independently promote cancer cell proliferation by reducing checkpoint-induced cell-cycle arrest during interphase. The co–up-regulation of RepoMan and Aurora B in tumors is inversely correlated with patient survival, underscoring its potential importance for tumor progression. Finally, we demonstrate that high RepoMan levels sensitize cancer cells to Aurora-B inhibitors. Hence, the co–up-regulation of RepoMan and Aurora B is associated with tumor aggressiveness but also exposes a vulnerable target for therapeutic intervention.
Collapse
Affiliation(s)
| | - Jan Rombouts
- Laboratory of Dynamics in Biological Systems, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium
| | - Mikhail Steklov
- VIB-KU Leuven Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Lorenzo Pasquali
- Dermatology and Venereology Section, Department of Medicine Solna, Karolinska Institutet, SE-17176 Stockholm, Sweden
| | - Anna Sablina
- Department of Oncology, KU Leuven, B-3000 Leuven, Belgium.,VIB-KU Leuven Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Lendert Gelens
- Laboratory of Dynamics in Biological Systems, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium
| | - Junbin Qian
- Laboratory of Biosignaling & Therapeutics, KU Leuven, B-3000 Leuven, Belgium.,Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, B-3000 Leuven, Belgium.,VIB-KU Leuven Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Mathieu Bollen
- Laboratory of Biosignaling & Therapeutics, KU Leuven, B-3000 Leuven, Belgium
| |
Collapse
|
32
|
Sami SA, Darwish NHE, Barile ANM, Mousa SA. Current and Future Molecular Targets for Acute Myeloid Leukemia Therapy. Curr Treat Options Oncol 2020; 21:3. [PMID: 31933183 DOI: 10.1007/s11864-019-0694-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OPINION STATEMENT Acute myeloid leukemia (AML) disease prognosis is poor and there is a high risk of chemo-resistant relapse for both young and old patients. Thus, there is a demand for alternative and target-specific drugs to improve the 5-year survival rate. Current treatment mainstays include chemotherapy, or mutation-specific targeting molecules including FLT3 inhibitors, IDH inhibitors, and monoclonal antibodies. Efforts to devise new, targeted therapy have included recent advances in methods for high-throughput genomic screening and the availability of computer-assisted techniques for the design of novel agents predicted to specifically inhibit mutant molecules involved in leukemogenesis. Crosstalk between the leukemia cells and the bone marrow microenvironment through cell surface molecules, such as the integrins αvβ3 and αvβ5, might influence drug response and AML progression. This review article focuses on current AML treatment options, new AML targeted therapies, the role of integrins in AML progression, and a potential therapeutic agent-integrin αvβ3 antagonist.
Collapse
Affiliation(s)
- Shaheedul A Sami
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, USA
| | - Noureldien H E Darwish
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, USA.,Hematology Unit, Clinical Pathology Department, Mansoura Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amanda N M Barile
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, USA
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, USA.
| |
Collapse
|
33
|
Ghelli Luserna di Rorà A, Martinelli G, Simonetti G. The balance between mitotic death and mitotic slippage in acute leukemia: a new therapeutic window? J Hematol Oncol 2019; 12:123. [PMID: 31771633 PMCID: PMC6880427 DOI: 10.1186/s13045-019-0808-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/17/2019] [Indexed: 12/11/2022] Open
Abstract
Mitosis is the process whereby an eukaryotic cell divides into two identical copies. Different multiprotein complexes are involved in the fine regulation of cell division, including the mitotic promoting factor and the anaphase promoting complex. Prolonged mitosis can result in cellular division, cell death, or mitotic slippage, the latter leading to a new interphase without cellular division. Mitotic slippage is one of the causes of genomic instability and has an important therapeutic and clinical impact. It has been widely studied in solid tumors but not in hematological malignancies, in particular, in acute leukemia. We review the literature data available on mitotic regulation, alterations in mitotic proteins occurring in acute leukemia, induction of prolonged mitosis and its consequences, focusing in particular on the balance between cell death and mitotic slippage and on its therapeutic potentials. We also present the most recent preclinical and clinical data on the efficacy of second-generation mitotic drugs (CDK1-Cyclin B1, APC/CCDC20, PLK, Aurora kinase inhibitors). Despite the poor clinical activity showed by these drugs as single agents, they offer a potential therapeutic window for synthetic lethal combinations aimed to selectively target leukemic cells at the right time, thus decreasing the risk of mitotic slippage events.
Collapse
Affiliation(s)
- Andrea Ghelli Luserna di Rorà
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy.
| | - Giovanni Martinelli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| | - Giorgia Simonetti
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, FC, Italy
| |
Collapse
|
34
|
Yuan T, Qi B, Jiang Z, Dong W, Zhong L, Bai L, Tong R, Yu J, Shi J. Dual FLT3 inhibitors: Against the drug resistance of acute myeloid leukemia in recent decade. Eur J Med Chem 2019; 178:468-483. [PMID: 31207462 DOI: 10.1016/j.ejmech.2019.06.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/16/2019] [Accepted: 06/02/2019] [Indexed: 01/18/2023]
Abstract
Acute myeloid leukemia (AML) is a malignant disease characterized by abnormal growth and differentiation of hematopoietic stem cells. Although the pathogenesis has not been fully elucidated, many specific gene mutations have been found in AML. Fms-like tyrosine kinase 3 (FLT3) is recognized as a drug target for the treatment of AML, and the activation mutations of FLT3 were found in about 30% of AML patients. Targeted inhibition of FLT3 receptor tyrosine kinase has shown promising results in the treatment of FLT3 mutation AML. Unfortunately, the therapeutic effects of FLT3 tyrosine kinase inhibitors used as AML monotherapy are usually accompanied by the high risk of resistance development within a few months after treatment. FLT3 dual inhibitors were generated with the co-inhibition of FLT3 and another target, such as CDK4, JAK2, MEK, Mer, Pim, etc., to solve the problems mentioned above. As a result, the therapeutic effect of the drug is significantly improved, while the toxic and side effects are reduced. Besides, the life quality of AML patients with FLT3 mutation has been effectively improved. In this paper, we reviewed the studies of dual FLT3 inhibitors that have been discovered in recent years for the treatment of AML.
Collapse
Affiliation(s)
- Ting Yuan
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Baowen Qi
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Zhongliang Jiang
- Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
| | - Wenjuan Dong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lei Zhong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lan Bai
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Rongsheng Tong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jiying Yu
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
35
|
Wang S, Hwang EE, Guha R, O'Neill AF, Melong N, Veinotte CJ, Conway Saur A, Wuerthele K, Shen M, McKnight C, Alexe G, Lemieux ME, Wang A, Hughes E, Xu X, Boxer MB, Hall MD, Kung A, Berman JN, Davis MI, Stegmaier K, Crompton BD. High-throughput Chemical Screening Identifies Focal Adhesion Kinase and Aurora Kinase B Inhibition as a Synergistic Treatment Combination in Ewing Sarcoma. Clin Cancer Res 2019; 25:4552-4566. [PMID: 30979745 DOI: 10.1158/1078-0432.ccr-17-0375] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 12/18/2018] [Accepted: 04/09/2019] [Indexed: 12/27/2022]
Abstract
PURPOSE Ewing sarcoma is an aggressive solid tumor malignancy of childhood. Although current treatment regimens cure approximately 70% of patients with localized disease, they are ineffective for most patients with metastases or relapse. New treatment combinations are necessary for these patients. EXPERIMENTAL DESIGN Ewing sarcoma cells are dependent on focal adhesion kinase (FAK) for growth. To identify candidate treatment combinations for Ewing sarcoma, we performed a small-molecule library screen to identify compounds synergistic with FAK inhibitors in impairing Ewing cell growth. The activity of a top-scoring class of compounds was then validated across multiple Ewing cell lines in vitro and in multiple xenograft models of Ewing sarcoma. RESULTS Numerous Aurora kinase inhibitors scored as synergistic with FAK inhibition in this screen. We found that Aurora kinase B inhibitors were synergistic across a larger range of concentrations than Aurora kinase A inhibitors when combined with FAK inhibitors in multiple Ewing cell lines. The combination of AZD-1152, an Aurora kinase B-selective inhibitor, and PF-562271 or VS-4718, FAK-selective inhibitors, induced apoptosis in Ewing sarcoma cells at concentrations that had minimal effects on survival when cells were treated with either drug alone. We also found that the combination significantly impaired tumor progression in multiple xenograft models of Ewing sarcoma. CONCLUSIONS FAK and Aurora kinase B inhibitors synergistically impair Ewing sarcoma cell viability and significantly inhibit tumor progression. This study provides preclinical support for the consideration of a clinical trial testing the safety and efficacy of this combination for patients with Ewing sarcoma.
Collapse
Affiliation(s)
- Sarah Wang
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Elizabeth E Hwang
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Rajarshi Guha
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Allison F O'Neill
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | | | - Chansey J Veinotte
- IWK Health Centre, Halifax, Nova Scotia, Canada
- Dalhousie University, Halifax, Nova Scotia, Canada
| | - Amy Conway Saur
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Kellsey Wuerthele
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Crystal McKnight
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Gabriela Alexe
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
- Boston University Bioinformatics Graduate Program, Boston, Massachusetts
| | | | - Amy Wang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Emma Hughes
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Xin Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Matthew B Boxer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Andrew Kung
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jason N Berman
- IWK Health Centre, Halifax, Nova Scotia, Canada
- Dalhousie University, Halifax, Nova Scotia, Canada
| | - Mindy I Davis
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Kimberly Stegmaier
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts.
| | - Brian D Crompton
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts.
| |
Collapse
|
36
|
Moison C, Lavallée VP, Thiollier C, Lehnertz B, Boivin I, Mayotte N, Gareau Y, Fréchette M, Blouin-Chagnon V, Corneau S, Lavallée S, Lemieux S, Marinier A, Hébert J, Sauvageau G. Complex karyotype AML displays G2/M signature and hypersensitivity to PLK1 inhibition. Blood Adv 2019; 3:552-563. [PMID: 30782614 PMCID: PMC6391664 DOI: 10.1182/bloodadvances.2018028480] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 01/11/2019] [Indexed: 01/07/2023] Open
Abstract
Patients diagnosed with acute myeloid leukemia with complex karyotype (CK AML) have an adverse prognosis using current therapies, especially when accompanied by TP53 alterations. We hereby report the RNA-sequencing analysis of the 68 CK AML samples included in the Leucegene 415 patient cohort. We confirm the frequent occurrence of TP53 alterations in this subgroup and further characterize the allele expression profile and transcript alterations of this gene. We also document that the RAS pathway (N/KRAS, NF1, PTPN11, BRAF) is frequently altered in this disease. Targeted chemical interrogation of genetically characterized primary CK AML samples identifies polo-like kinase 1 (PLK1) inhibitors as the most selective agents for this disease subgroup. TP53 status did not alter sensitivity to PLK1 inhibitors. Interestingly, CK AML specimens display a G2/M transcriptomic signature that includes higher expression levels of PLK1 and correlates with PLK1 inhibition sensitivity. Together, our results highlight vulnerability in CK AML. In line with these in vitro data, volasertib shows a strong anti-AML activity in xenotransplantation mouse models of human adverse AML. Considering that PLK1 inhibitors are currently being investigated clinically in AML and myelodysplastic syndromes, our results provide a new rationale for PLK1-directed therapy in patients with adverse cytogenetic AML.
Collapse
Affiliation(s)
- Céline Moison
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Vincent-Philippe Lavallée
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Division of Hematology, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada
| | - Clarisse Thiollier
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Bernhard Lehnertz
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Isabel Boivin
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Nadine Mayotte
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Yves Gareau
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Department of Chemistry, Université de Montréal, Montréal, QC, Canada
| | - Mélanie Fréchette
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Valérie Blouin-Chagnon
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Sophie Corneau
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Sylvie Lavallée
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada; and
| | - Sébastien Lemieux
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Department of Computer Science and Operations Research and
| | - Anne Marinier
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Department of Chemistry, Université de Montréal, Montréal, QC, Canada
| | - Josée Hébert
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Division of Hematology, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada
- Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada; and
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Guy Sauvageau
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Division of Hematology, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada
- Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada; and
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
37
|
Liu X, Chen Y, Li Y, Petersen RB, Huang K. Targeting mitosis exit: A brake for cancer cell proliferation. Biochim Biophys Acta Rev Cancer 2019; 1871:179-191. [PMID: 30611728 DOI: 10.1016/j.bbcan.2018.12.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/03/2018] [Accepted: 12/03/2018] [Indexed: 12/16/2022]
Abstract
The transition from mitosis to interphase, referred to as mitotic exit, is a critical mitotic process which involves activation and inactivation of multiple mitotic kinases and counteracting protein phosphatases. Loss of mitotic exit checkpoints is a common feature of cancer cells, leading to mitotic dysregulation and confers cancer cells with oncogenic characteristics, such as aberrant proliferation and microtubule-targeting agent (MTA) resistance. Since MTA resistance results from cancer cells prematurely exiting mitosis (mitotic slippage), blocking mitotic exit is believed to be a promising anticancer strategy. Moreover, based on this theory, simultaneous inhibition of mitotic exit and additional cell cycle phases would likely achieve synergistic antitumor effects. In this review, we divide the molecular regulators of mitotic exit into four categories based on their different regulatory functions: 1) the anaphase-promoting complex/cyclosome (APC/C, a ubiquitin ligase), 2) cyclin B, 3) mitotic kinases and phosphatases, 4) kinesins and microtubule-binding proteins. We also review the regulators of mitotic exit and propose prospective anticancer strategies targeting mitotic exit, including their strengths and possible challenges to their use.
Collapse
Affiliation(s)
- Xinran Liu
- Tongji School of Pharmacy, Huazhong University of Science & Technology, Wuhan, Hubei 430030, China
| | - Yuchen Chen
- Tongji School of Pharmacy, Huazhong University of Science & Technology, Wuhan, Hubei 430030, China
| | - Yangkai Li
- Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Robert B Petersen
- Foundational Sciences, Central Michigan University College of Medicine, Mt. Pleasant, MI 48858, USA
| | - Kun Huang
- Tongji School of Pharmacy, Huazhong University of Science & Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
38
|
Payton M, Cheung HK, Ninniri MSS, Marinaccio C, Wayne WC, Hanestad K, Crispino JD, Juan G, Coxon A. Dual Targeting of Aurora Kinases with AMG 900 Exhibits Potent Preclinical Activity Against Acute Myeloid Leukemia with Distinct Post-Mitotic Outcomes. Mol Cancer Ther 2018; 17:2575-2585. [PMID: 30266802 PMCID: PMC6279493 DOI: 10.1158/1535-7163.mct-18-0186] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/15/2018] [Accepted: 09/25/2018] [Indexed: 01/19/2023]
Abstract
Aurora kinase A and B have essential and non-overlapping roles in mitosis, with elevated expression in a subset of human cancers, including acute myeloid leukemia (AML). In this study, pan-aurora kinase inhibitor (AKI) AMG 900 distinguishes itself as an anti-leukemic agent that is more uniformly potent against a panel of AML cell lines than are isoform-selective AKIs and classic AML drugs. AMG 900 inhibited AML cell growth by inducing polyploidization and/or apoptosis. AMG 900 and aurora-B-selective inhibitor AZD1152-hQPA showed comparable cellular effects on AML lines that do not harbor a FLT3-ITD mutation. AMG 900 was active against P-glycoprotein-expressing AML cells resistant to AZD1152-hQPA and was effective at inducing expression of megakaryocyte-lineage markers (CD41, CD42) on human CHRF-288-11 cells and mouse Jak2 V617F cells. In MOLM-13 cells, inhibition of p-histone H3 by AMG 900 was associated with polyploidy, extra centrosomes, accumulation of p53 protein, apoptosis, and cleavage of Bcl-2 protein. Co-administration of cytarabine (Ara-C) with AMG 900 potentiated cell killing in a subset of AML lines, with evidence of attenuated polyploidization. AMG 900 inhibited the proliferation of primary human bone marrow cells in culture, with a better proliferation recovery profile relative to classic antimitotic drug docetaxel. In vivo, AMG 900 significantly reduced tumor burden in a systemic MOLM-13 xenograft model where we demonstrate the utility of 3'-deoxy-3'-18F-fluorothymidine [18F]FLT positron emission tomographic (PET)-CT imaging to measure the antiproliferative effects of AMG 900 in skeletal tissues in mice.
Collapse
Affiliation(s)
- Marc Payton
- Amgen Discovery Research, Thousand Oaks, California.
| | | | | | | | | | | | - John D Crispino
- Division of Hematology/Oncology, Northwestern University, Chicago, Illinois
| | - Gloria Juan
- Amgen Medical Sciences, Thousand Oaks, California
| | - Angela Coxon
- Amgen Discovery Research, Thousand Oaks, California
| |
Collapse
|
39
|
Henriques AC, Ribeiro D, Pedrosa J, Sarmento B, Silva PMA, Bousbaa H. Mitosis inhibitors in anticancer therapy: When blocking the exit becomes a solution. Cancer Lett 2018; 440-441:64-81. [PMID: 30312726 DOI: 10.1016/j.canlet.2018.10.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/12/2018] [Accepted: 10/02/2018] [Indexed: 12/11/2022]
Abstract
Current microtubule-targeting agents (MTAs) remain amongst the most important antimitotic drugs used against a broad range of malignancies. By perturbing spindle assembly, MTAs activate the spindle assembly checkpoint (SAC), which induces mitotic arrest and subsequent apoptosis. However, besides toxic side effects and resistance, mitotic slippage and failure in triggering apoptosis in various cancer cells are limiting factors of MTAs efficacy. Alternative strategies to target mitosis without affecting microtubules have, thus, led to the identification of small molecules, such as those that target spindle Kinesins, Aurora and Polo-like kinases. Unfortunately, these so-called second-generation of antimitotics, encompassing mitotic blockers and mitotic drivers, have failed in clinical trials. Our recent understanding regarding the mechanisms of cell death during a mitotic arrest pointed out apoptosis as the main variable, providing an opportunity to control the cell fates and influence the effectiveness of antimitotics. Here, we provide an overview on the second-generation of antimitotics, and discuss possible strategies that exploit SAC activity, mitotic slippage/exit and apoptosis induction, in order to improve the efficacy of anticancer strategies that target mitosis.
Collapse
Affiliation(s)
- Ana C Henriques
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal; INEB, Instituto Nacional de Engenharia Biomédica, Universidade Do Porto, Porto, Portugal
| | - Diana Ribeiro
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal; Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR/CIMAR), Universidade Do Porto, Porto, Portugal
| | - Joel Pedrosa
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal
| | - Bruno Sarmento
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal; INEB, Instituto Nacional de Engenharia Biomédica, Universidade Do Porto, Porto, Portugal; i3S - Instituto de Investigação e Inovação Em Saúde, Universidade Do Porto, Porto, Portugal
| | - Patrícia M A Silva
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal
| | - Hassan Bousbaa
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal; Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR/CIMAR), Universidade Do Porto, Porto, Portugal.
| |
Collapse
|
40
|
Noronha S, Alt LAC, Scimeca TE, Zarou O, Obrzut J, Zanotti B, Hayward EA, Pillai A, Mathur S, Rojas J, Salamah R, Chandar N, Fay MJ. Preclinical evaluation of the Aurora kinase inhibitors AMG 900, AZD1152-HQPA, and MK-5108 on SW-872 and 93T449 human liposarcoma cells. In Vitro Cell Dev Biol Anim 2017; 54:71-84. [PMID: 29197031 DOI: 10.1007/s11626-017-0208-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 10/10/2017] [Indexed: 11/26/2022]
Abstract
Liposarcoma is a malignant soft tissue tumor that originates from adipose tissue and is one of the most frequently diagnosed soft tissue sarcomas in humans. There is great interest in identifying novel chemotherapeutic options for treating liposarcoma based upon molecular alterations in the cancer cells. The Aurora kinases have been identified as promising chemotherapeutic targets based on their altered expression in many human cancers and cellular roles in mitosis and cytokinesis. In this study, we investigated the effects of an Aurora kinase A inhibitor (MK-5108), an Aurora kinase B inhibitor (AZD1152-HQPA), and a pan-Aurora kinase inhibitor (AMG 900) on undifferentiated SW-872 and well-differentiated 93T449 human liposarcoma cells. Treatment of the SW-872 and 93T449 cells with MK-5108 (0-1000 nM), AZD1152-HQPA (0-1000 nM), and AMG 900 (0-1000 nM) for 72 h resulted in a dose-dependent decrease in the total viable cell number. Based upon the EC50 values, the potency of the three Aurora kinase inhibitors in the SW-872 cells was as follows: AMG 900 (EC50 = 3.7 nM) > AZD1152-HQPA (EC50 = 43.4 nM) > MK-5108 (EC50 = 309.0 nM), while the potency in the 93T449 cells was as follows: AMG 900 (EC50 = 6.5 nM) > AZD1152-HQPA (EC50 = 74.5 nM) > MK-5108 (EC50 = 283.6 nM). The percentage of polyploidy after 72 h of drug treatment (0-1000 nM) was determined by propidium iodide staining and flow cytometric analysis. AMG 900 caused a significant increase in polyploidy starting at 25 nM in the SW-872 and 93T449 cells, and AZD1152-HQPA caused a significant increase starting at 100 nM in the SW-872 cells and 250 nM in the 93T449 cells. The Aurora kinase A inhibitor MK-5108 did not significantly increase the percentage of polyploid cells at any of the doses tested in either cell line. The expression of Aurora kinase A and B was evaluated in the SW-872 cells versus differentiated adipocytes and human mesenchymal stem cells by real-time RT-PCR and Western blot analysis. Aurora kinase A and B mRNA expression was significantly increased in the SW-872 cells versus the differentiated adipocytes and human mesenchymal stem cells. Western blot analysis revealed a ~ 48 kDa immunoreactive band for Aurora kinase A that was not present in the differentiated adipocytes or the human mesenchymal stem cells. A ~ 39 kDa immunoreactive band for Aurora kinase B was detected in the SW-872 cells, differentiated adipocytes, and human mesenchymal stem cells. A smaller immunoreactive band for Aurora kinase B was detected in the SW-872 cells but not in the differentiated adipocytes and human mesenchymal stem cells, and this may reflect the expression of a truncated splice variant of Aurora kinase B that has been associated with poor patient prognosis. The 93T449 cells demonstrated decreased expression of Aurora kinase A and B mRNA and protein compared to the SW-872 cells, and also expressed the truncated form of Aurora kinase B. The results of these in vitro studies indicate that Aurora kinase inhibitors should be further investigated as possible chemotherapeutic agents for human liposarcoma.
Collapse
Affiliation(s)
- Sandhya Noronha
- Physician Assistant Program, College of Health Sciences, Midwestern University, 555 31st Street, Downers Grove, IL, 60515, USA
| | - Lauren A C Alt
- Department of Biomedical Sciences, College of Health Sciences, Midwestern University, 555 31st Street, Downers Grove, IL, 60515, USA
| | - Taylor E Scimeca
- Department of Biomedical Sciences, College of Health Sciences, Midwestern University, 555 31st Street, Downers Grove, IL, 60515, USA
| | - Omran Zarou
- Department of Biomedical Sciences, College of Health Sciences, Midwestern University, 555 31st Street, Downers Grove, IL, 60515, USA
| | - Justyna Obrzut
- Department of Biomedical Sciences, College of Health Sciences, Midwestern University, 555 31st Street, Downers Grove, IL, 60515, USA
| | - Brian Zanotti
- Department of Microbiology and Immunology, Chicago College of Osteopathic Medicine, Midwestern University, 555 31st Street, Downers Grove, IL, 60515, USA
| | - Elizabeth A Hayward
- Department of Biomedical Sciences, College of Health Sciences, Midwestern University, 555 31st Street, Downers Grove, IL, 60515, USA
| | - Akhil Pillai
- Department of Pharmacology, Chicago College of Osteopathic Medicine, Midwestern University, 555 31st Street, Downers Grove, IL, 60515, USA
| | - Shubha Mathur
- Department of Pharmacology, Chicago College of Osteopathic Medicine, Midwestern University, 555 31st Street, Downers Grove, IL, 60515, USA
| | - Joseph Rojas
- Department of Biomedical Sciences, College of Health Sciences, Midwestern University, 555 31st Street, Downers Grove, IL, 60515, USA
| | - Ribhi Salamah
- Department of Biomedical Sciences, College of Health Sciences, Midwestern University, 555 31st Street, Downers Grove, IL, 60515, USA
| | - Nalini Chandar
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, 555 31st Street, Downers Grove, IL, 60515, USA
| | - Michael J Fay
- Department of Biomedical Sciences, College of Health Sciences, Midwestern University, 555 31st Street, Downers Grove, IL, 60515, USA.
- Department of Pharmacology, Chicago College of Osteopathic Medicine, Midwestern University, 555 31st Street, Downers Grove, IL, 60515, USA.
| |
Collapse
|
41
|
Tang A, Gao K, Chu L, Zhang R, Yang J, Zheng J. Aurora kinases: novel therapy targets in cancers. Oncotarget 2017; 8:23937-23954. [PMID: 28147341 PMCID: PMC5410356 DOI: 10.18632/oncotarget.14893] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/17/2017] [Indexed: 02/06/2023] Open
Abstract
Aurora kinases, a family of serine/threonine kinases, consisting of Aurora A (AURKA), Aurora B (AURKB) and Aurora C (AURKC), are essential kinases for cell division via regulating mitosis especially the process of chromosomal segregation. Besides regulating mitosis, Aurora kinases have been implicated in regulating meiosis. The deletion of Aurora kinases could lead to failure of cell division and impair the embryonic development. Overexpression or gene amplification of Aurora kinases has been clarified in a number of cancers. And a growing number of studies have demonstrated that inhibition of Aurora kinases could potentiate the effect of chemotherapies. For the past decades, a series of Aurora kinases inhibitors (AKIs) developed effectively repress the progression and growth of many cancers both in vivo and in vitro, suggesting that Aurora kinases could be a novel therapeutic target. In this review, we'll first briefly present the structure, localization and physiological functions of Aurora kinases in mitosis, then describe the oncogenic role of Aurora kinases in tumorigenesis, we shall finally discuss the outcomes of AKIs combination with conventional therapy.
Collapse
Affiliation(s)
- Anqun Tang
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China
| | - Keyu Gao
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China
| | - Laili Chu
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China
| | - Rui Zhang
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China
| | - Jing Yang
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China
| | - Junnian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China.,Department of Oncology, The First Affiliated Hospital, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
42
|
Ferguson FM, Doctor ZM, Chaikuad A, Sim T, Kim ND, Knapp S, Gray NS. Characterization of a highly selective inhibitor of the Aurora kinases. Bioorg Med Chem Lett 2017; 27:4405-4408. [PMID: 28818446 PMCID: PMC11892687 DOI: 10.1016/j.bmcl.2017.08.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/08/2017] [Accepted: 08/09/2017] [Indexed: 11/23/2022]
Abstract
Aurora kinases play an essential role in mitosis and cell cycle regulation. In recent years Aurora kinases have proved popular cancer targets and many inhibitors have been developed. The majority of these clinical candidates are multi-targeted, rendering them inappropriate as tools for studying Aurora kinase mediated signaling. Here we report discovery of a highly selective inhibitor of Aurora kinases A, B and C, with potent cellular activity and minimal off-target activity (PLK4). The X-ray co-crystal structure of Aurora A in complex with compound 2 is reported, and provides insights into the structural determinants of ligand binding and selectivity.
Collapse
Affiliation(s)
- Fleur M Ferguson
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Zainab M Doctor
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Apirat Chaikuad
- Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom; Institute for Pharmaceutical Chemistry and Buchmann Institute for Life Sciences, Goethe University, Max-von Laue Str. 9, 60438 Frankfurt am Main, Germany
| | - Taebo Sim
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Republic of Korea
| | - Nam Doo Kim
- Daegu-Gyeongbuk Medical Innovation Foundation, Republic of Korea
| | - Stefan Knapp
- Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom; German Cancer Consortium (DKTK), Frankfurt site, Germany; Institute for Pharmaceutical Chemistry and Buchmann Institute for Life Sciences, Goethe University, Max-von Laue Str. 9, 60438 Frankfurt am Main, Germany
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
43
|
Kantarjian HM, Schuster MW, Jain N, Advani A, Jabbour E, Gamelin E, Rasmussen E, Juan G, Anderson A, Chow VF, Friberg G, Vogl FD, Sekeres MA. A phase 1 study of AMG 900, an orally administered pan-aurora kinase inhibitor, in adult patients with acute myeloid leukemia. Am J Hematol 2017; 92:660-667. [PMID: 28370201 PMCID: PMC5925751 DOI: 10.1002/ajh.24736] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/20/2017] [Accepted: 03/21/2017] [Indexed: 01/05/2023]
Abstract
Aurora kinases are involved in the pathophysiology of several cancers including acute myeloid leukemia (AML). In this phase 1 study, we investigated the safety and efficacy of AMG 900, an orally administered, highly potent, selective, small-molecule inhibitor of both Aurora kinase A and B, in patients with AML . Patients with pathologically documented AML who either declined standard treatments or had relapsed from or were refractory to previous therapies were enrolled. Two every-2-week dose-escalation schedules using a modified 3 + 3 + 3 design were evaluated AMG 900 given daily for 4 days with 10 days off (4/10 schedule), and AMG 900 given daily for 7 days with 7 days off (7/7 schedule). Thirty-five patients were enrolled at 9 different dose levels: 22 patients on the 4/10 schedule (doses from 15 to 100 mg daily), and 13 patients on the 7/7 schedule (doses from 30 to 50 mg daily). Both schedules were tolerated; nausea (31%), diarrhea (29%), febrile neutropenia (29%), and fatigue (23%) were the most common treatment-related adverse events. Three patients (9%) achieved complete response with incomplete count recovery. Patients with higher baseline expression of a set of specific pathway-related genes (BIRC5, AURKA, TTK, CDC2, and CCNB1) were more likely to respond in an exploratory biomarker analysis. AMG 900 was tolerated in a general AML population, and pathway-specific biomarkers identified a potential target population. Future research efforts will be directed toward further exploration of biomarkers of response and combination of AMG 900 with other anticancer agents.
Collapse
Affiliation(s)
- Hagop M. Kantarjian
- Department of Leukemia, MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | - Michael W. Schuster
- Medical Hematology/Oncology Stony Brook University School of Medicine, Stony Brook, New York, USA
| | - Nitin Jain
- Department of Leukemia, MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | - Anjali Advani
- Leukemia Program, Cleveland Clinic, Cleveland, Ohio, USA
| | - Elias Jabbour
- Department of Leukemia, MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | - Erick Gamelin
- Research and Development, Amgen Inc., Thousand Oaks, California, USA
| | - Erik Rasmussen
- Research and Development, Amgen Inc., Thousand Oaks, California, USA
| | - Gloria Juan
- Research and Development, Amgen Inc., Thousand Oaks, California, USA
| | - Abraham Anderson
- Research and Development, Amgen Inc., Thousand Oaks, California, USA
| | - Vincent F. Chow
- Research and Development, Amgen Inc., Thousand Oaks, California, USA
| | - Gregory Friberg
- Research and Development, Amgen Inc., Thousand Oaks, California, USA
| | - Florian D. Vogl
- Research and Development, Amgen Inc., Thousand Oaks, California, USA
| | | |
Collapse
|
44
|
Abstract
INTRODUCTION AML therapy remains very challenging despite our increased understanding of its molecular heterogeneity. Outcomes with chemotherapy and targeted therapy remain poor. Targeting cell cycle regulators might complement chemotherapy and targeted therapy and help in improving outcomes. Areas covered: Here we cover the pre-clinical and clinical data for both for cyclin dependent kinase (CDK) and cell-cycle checkpoint inhibitors. While CDK inhibition can inhibit proliferation, checkpoint inhibitors can facilitate cell cycle progression in presence of DNA damage and can induce mitotic catastrophe. Expert opinion: Though the preclinical data for cell cycle inhibitors in AML is compelling, the clinical translation so far has proven to be challenging. This is a reflection of the complexity of both, AML and cell cycle regulators. However, early introduction of cell-cycle active agents in combination with chemotherapy or targeted agents, identifying right sequence of use and identifying right biomarkers might pave the way into successful clinical translation.
Collapse
Affiliation(s)
- Abdallah Abou Zahr
- a Department of Leukemia , University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Gautam Borthakur
- a Department of Leukemia , University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
45
|
Abstract
Mutations in cancer cells frequently result in cell cycle alterations that lead to unrestricted growth compared to normal cells. Considering this phenomenon, many drugs have been developed to inhibit different cell-cycle phases. Mitotic phase targeting disturbs mitosis in tumor cells, triggers the spindle assembly checkpoint and frequently results in cell death. The first anti-mitotics to enter clinical trials aimed to target tubulin. Although these drugs improved the treatment of certain cancers, and many anti-microtubule compounds are already approved for clinical use, severe adverse events such as neuropathies were observed. Since then, efforts have been focused on the development of drugs that also target kinases, motor proteins and multi-protein complexes involved in mitosis. In this review, we summarize the major proteins involved in the mitotic phase that can also be targeted for cancer treatment. Finally, we address the activity of anti-mitotic drugs tested in clinical trials in recent years.
Collapse
|
46
|
Olziersky AM, Labidi-Galy SI. Clinical Development of Anti-mitotic Drugs in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1002:125-152. [PMID: 28600785 DOI: 10.1007/978-3-319-57127-0_6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mitosis is one of the most fundamental processes of life by which a mammalian cell divides into two daughter cells. Mitosis has been an attractive target for anticancer therapies since fast proliferation was identified as one of the hallmarks of cancer cells. Despite efforts into developing specific inhibitors for mitotic kinases and kinesins, very few drugs have shown the efficiency of microtubule targeting-agents in cancer cells with paclitaxel being the most successful. A deeper translational research accompanying clinical trials of anti-mitotic drugs will help in identifying potent biomarkers predictive for response. Here, we review the current knowledge of mitosis targeting agents that have been tested so far in the clinics.
Collapse
Affiliation(s)
- Anna-Maria Olziersky
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - S Intidhar Labidi-Galy
- Department of Oncology, Geneva University Hospitals, Rue Gabrielle Perret-Gentil 4, Geneva, 1205, Switzerland.
| |
Collapse
|
47
|
Bogen D, Wei JS, Azorsa DO, Ormanoglu P, Buehler E, Guha R, Keller JM, Mathews Griner LA, Ferrer M, Song YK, Liao H, Mendoza A, Gryder BE, Sindri S, He J, Wen X, Zhang S, Shern JF, Yohe ME, Taschner-Mandl S, Shohet JM, Thomas CJ, Martin SE, Ambros PF, Khan J. Aurora B kinase is a potent and selective target in MYCN-driven neuroblastoma. Oncotarget 2016; 6:35247-62. [PMID: 26497213 PMCID: PMC4742102 DOI: 10.18632/oncotarget.6208] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/30/2015] [Indexed: 01/11/2023] Open
Abstract
Despite advances in multimodal treatment, neuroblastoma (NB) is often fatal for children with high-risk disease and many survivors need to cope with long-term side effects from high-dose chemotherapy and radiation. To identify new therapeutic targets, we performed an siRNA screen of the druggable genome combined with a small molecule screen of 465 compounds targeting 39 different mechanisms of actions in four NB cell lines. We identified 58 genes as targets, including AURKB, in at least one cell line. In the drug screen, aurora kinase inhibitors (nine molecules) and in particular the AURKB-selective compound, barasertib, were the most discriminatory with regard to sensitivity for MYCN-amplified cell lines. In an expanded panel of ten NB cell lines, those with MYCN-amplification and wild-type TP53 were the most sensitive to low nanomolar concentrations of barasertib. Inhibition of the AURKB kinase activity resulted in decreased phosphorylation of the known target, histone H3, and upregulation of TP53 in MYCN-amplified, TP53 wild-type cells. However, both wild-type and TP53 mutant MYCN-amplified cell lines arrested in G2/M phase upon AURKB inhibition. Additionally, barasertib induced endoreduplication and apoptosis. Treatment of MYCN-amplified/TP53 wild-type neuroblastoma xenografts resulted in profound growth inhibition and tumor regression. Therefore, aurora B kinase inhibition is highly effective in aggressive neuroblastoma and warrants further investigation in clinical trials.
Collapse
Affiliation(s)
- Dominik Bogen
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Jun S Wei
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - David O Azorsa
- Clinical Translational Research Division, Translational Genomics Research Institute (TGen), Scottsdale, AZ, USA
| | - Pinar Ormanoglu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Eugen Buehler
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Jonathan M Keller
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Lesley A Mathews Griner
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Young K Song
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hongling Liao
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arnulfo Mendoza
- Tumor and Metastasis Biology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Berkley E Gryder
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sivasish Sindri
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jianbin He
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xinyu Wen
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shile Zhang
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - John F Shern
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marielle E Yohe
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sabine Taschner-Mandl
- Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Jason M Shohet
- Texas Children's Cancer Center and Center for Cell and Gene Therapy, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Scott E Martin
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Peter F Ambros
- Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Javed Khan
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
48
|
PRMT1 promotes mitosis of cancer cells through arginine methylation of INCENP. Oncotarget 2016; 6:35173-82. [PMID: 26460953 PMCID: PMC4742097 DOI: 10.18632/oncotarget.6050] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 09/25/2015] [Indexed: 11/25/2022] Open
Abstract
Inner centromere protein (INCENP) is a part of a protein complex known as the chromosomal passenger complex (CPC) that is essential for correcting non-bipolar chromosome attachments and for cytokinesis. We here demonstrate that a protein arginine methyltransferase PRMT1, which are overexpressed in various types of cancer including lung and bladder cancer, methylates arginine 887 in an Aurora Kinase B (AURKB)-binding region of INCENP both in vitro and in vivo. R887-substituted INCENP revealed lower binding-affinity to AURKB than wild-type INCENP in the presence of PRMT1. Knockdown of PRMT1 as well as overexpression of methylation-inactive INCENP attenuated the AURKB activity in cancer cells, and resulted in abnormal chromosomal alignment and segregation. Furthermore, introduction of methylation-inactive INCENP into cancer cells reduced the growth rate, compared with those introduced wild-type INCENP or Mock. Our data unveils a novel mechanism of PRMT1-mediated CPC regulation through methylation of INCENP.
Collapse
|
49
|
Le LTT, Couvet M, Favier B, Coll JL, Nguyen CH, Molla A. Discovery of benzo[e]pyridoindolones as kinase inhibitors that disrupt mitosis exit while erasing AMPK-Thr172 phosphorylation on the spindle. Oncotarget 2016; 6:22152-66. [PMID: 26247630 PMCID: PMC4673153 DOI: 10.18632/oncotarget.4158] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/30/2015] [Indexed: 01/09/2023] Open
Abstract
Aurora kinases play an essential role in mitotic progression and are attractive targets in cancer therapy. The first generation of benzo[e]pyridoindole exhibited powerful aurora kinase inhibition but their low solubility limited further development. Grafting a pyperidine-ethoxy group gives rise to a hydrosoluble inhibitor: compound C5M.C5M could efficiently inhibit the proliferation of cells from different origins. C5M prevented cell cycling, induced a strong mitotic arrest then, cells became polyploid and finally died. C5M did not impair the spindle checkpoint, the separation of the sister chromatids and the transfer of aurora B on the mid-zone. C5M prevented histone H3 phosphorylation at mitotic entry and erased AMPK-Thr172 phosphorylation in late mitosis. With this unique profile of inhibition, C5M could be useful for understanding the role of phospho-Thr172-AMPK in abscission and the relationship between the chromosomal complex and the energy sensing machinery.C5M is a multikinase inhibitor with interesting preclinical characteristics: high hydro-solubility and a good stability in plasma. A single dose prevents the expansion of multicellular spheroids. C5M can safely be injected to mice and reduces significantly the development of xenograft. The next step will be to define the protocol of treatment and the cancer therapeutic field of this new anti-proliferative drug.
Collapse
Affiliation(s)
- Ly-Thuy-Tram Le
- INSERM UJF U823 Institut Albert Bonniot, Team 5, BP 170, Grenoble Cedex 9, France.,Department of Biotechnology, University of Sciences and Technology, DaNang, Vietnam
| | - Morgane Couvet
- INSERM UJF U823 Institut Albert Bonniot, Team 5, BP 170, Grenoble Cedex 9, France
| | - Bertrand Favier
- Université Joseph Fourier - Grenoble, Team GREPI, Etablissement Français du Sang, BP35, La Tronche France
| | - Jean-Luc Coll
- INSERM UJF U823 Institut Albert Bonniot, Team 5, BP 170, Grenoble Cedex 9, France
| | - Chi-Hung Nguyen
- Institut Curie, PSL Research University, UMR 9187 - U 1196 CNRS-Institut Curie, INSERM, Bat 110 Centre Universitaire, Orsay, France
| | - Annie Molla
- INSERM UJF U823 Institut Albert Bonniot, Team 5, BP 170, Grenoble Cedex 9, France
| |
Collapse
|
50
|
Cicenas J. The Aurora kinase inhibitors in cancer research and therapy. J Cancer Res Clin Oncol 2016; 142:1995-2012. [PMID: 26932147 DOI: 10.1007/s00432-016-2136-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 02/18/2016] [Indexed: 12/20/2022]
Abstract
Compounds that affect enzymatic function of kinases are valuable for the understanding of the complex biochemical processes in cells. Aurora kinases (AURKs) play a key role in the control of the mitosis. These kinases are frequently deregulated in different human cancers: overexpression, amplifications, translocations and deletions were reported in many cancer cell lines as well as patient tissues. These findings steered a rigorous hunt for small-molecule AURK inhibitors not only for research purposes as well as for therapeutic uses. In this review, we describe a number of AURK inhibitors and their use in cancer research and/or therapy. We hope to assist researchers and clinicians in deciding which inhibitor is most appropriate for their specific purpose. The review will also provide a broad overview of the clinical studies performed with some of these inhibitors (if such studies have been performed).
Collapse
Affiliation(s)
- Jonas Cicenas
- CALIPHO Group, Swiss Institute of Bioinformatics, CMU-1, rue Michel Servet, 1211, Geneva 4, Switzerland.
- MAP Kinase Resource, Melchiorstrasse 9, 3027, Bern, Switzerland.
- Proteomics Centre, Vilnius University Institute of Biochemistry, 08662, Vilnius, Lithuania.
| |
Collapse
|