1
|
Piastra V, Ganci F, Sacconi A, Pranteda A, Allegretti M, Bernardini R, Serra M, Lupo B, Dell'Aquila E, Ferretti G, Pescarmona E, Bartolazzi A, Blandino G, Trusolino L, Bossi G. Repurposed AT9283 triggers anti-tumoral effects by targeting MKK3 oncogenic functions in Colorectal Cancer. J Exp Clin Cancer Res 2024; 43:234. [PMID: 39164711 PMCID: PMC11334304 DOI: 10.1186/s13046-024-03150-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 08/04/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common type of cancer and the second leading cause of cancer-related deaths worldwide, with a survival rate near to 10% when diagnosed at an advanced stage. Hence, the identification of new molecular targets to design more selective and efficient therapies is urgently required. The Mitogen activated protein kinase kinase 3 (MKK3) is a dual-specificity threonine/tyrosine protein kinase that, activated in response to cellular stress and inflammatory stimuli, regulates a plethora of biological processes. Previous studies revealed novel MKK3 roles in supporting tumor malignancy, as its depletion induces autophagy and cell death in cancer lines of different tumor types, including CRC. Therefore, MKK3 may represent an interesting new therapeutic target in advanced CRC, however selective MKK3 inhibitors are currently not available. METHODS The study involved transcriptomic based drug repurposing approach and confirmatory assays with CRC lines, primary colonocytes and a subset of CRC patient-derived organoids (PDO). Investigations in vitro and in vivo were addressed. RESULTS The repurposing approach identified the multitargeted kinase inhibitor AT9283 as a putative compound with MKK3 depletion-mimicking activities. Indeed, AT9283 drops phospho- and total-MKK3 protein levels in tested CRC models. Likely the MKK3 silencing, AT9283 treatment: i) inhibited cell proliferation promoting autophagy and cell death in tested CRC lines and PDOs; ii) resulted well-tolerated by CCD-18Co colonocytes; iii) reduced cancer cell motility inhibiting CRC cell migration and invasion; iv) inhibited COLO205 xenograft tumor growth. Mechanistically, AT9283 abrogated MKK3 protein levels mainly through the inhibition of aurora kinase A (AURKA), impacting on MKK3/AURKA protein-protein interaction and protein stability therefore uncovering the relevance of MKK3/AURKA crosstalk in sustaining CRC malignancy in vitro and in vivo. CONCLUSION Overall, we demonstrated that the anti-tumoral effects triggered by AT9283 treatment recapitulated the MKK3 depletion effects in all tested CRC models in vitro and in vivo, suggesting that AT9283 is a repurposed drug. According to its good tolerance when tested with primary colonocytes (CCD-18CO), AT9283 is a promising drug for the development of novel therapeutic strategies to target MKK3 oncogenic functions in late-stage and metastatic CRC patients.
Collapse
Affiliation(s)
- Valentina Piastra
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy
- Department of Science, University Roma Tre, Rome, Italy
| | - Federica Ganci
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy
| | - Andrea Sacconi
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy
| | - Angelina Pranteda
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy
- Department of Science, University Roma Tre, Rome, Italy
| | - Matteo Allegretti
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy
| | - Roberta Bernardini
- Interdepartmental Center for Comparative Medicine, Alternative Techniques and Aquaculture (CIMETA), University of Rome Tor Vergata, Rome, Italy
| | - Martina Serra
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy
- Interdepartmental Center for Comparative Medicine, Alternative Techniques and Aquaculture (CIMETA), University of Rome Tor Vergata, Rome, Italy
| | - Barbara Lupo
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Candiolo, Turin, Italy
| | - Emanuela Dell'Aquila
- Second Division of Medical Oncology, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Gianluigi Ferretti
- First Division of Medical Oncology, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Edoardo Pescarmona
- Department of Pathology, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Armando Bartolazzi
- Pathology Research Laboratory, St Andrea University Hospital, Rome, Italy
| | - Giovanni Blandino
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy
| | - Livio Trusolino
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Candiolo, Turin, Italy
| | - Gianluca Bossi
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy.
| |
Collapse
|
2
|
Gupta D, Kumar M, Saifi S, Rawat S, Ethayathulla AS, Kaur P. A comprehensive review on role of Aurora kinase inhibitors (AKIs) in cancer therapeutics. Int J Biol Macromol 2024; 265:130913. [PMID: 38508544 DOI: 10.1016/j.ijbiomac.2024.130913] [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: 11/07/2023] [Revised: 03/09/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024]
Abstract
Aurora kinases (AURKs) are a family of serine /threonine protein kinases that have a crucial role in cell cycle process mainly in the event of chromosomal segregation, centrosome maturation and cytokinesis. The family consists of three members including Aurora kinase A (AURK-A), Aurora kinase B (AURK-B) and Aurora kinase C (AURK-C). All AURKs contain a conserved kinase domain for their activity but differ in their cellular localization and functions. AURK-A and AURK-B are expressed mainly in somatic cells while the expression of AURK-C is limited to germ cells. AURK-A promotes G2 to M transition of cell cycle by controlling centrosome maturation and mitotic spindle assembly. AURK-B and AURK-C form the chromosome passenger complex (CPC) that ensures proper chromosomal alignments and segregation. Aberrant expression of AURK-A and AURK-B has been detected in several solid tumours and malignancies. Hence, they have become an attractive therapeutic target against cancer. The first part of this review focuses on AURKs structure, functions, subcellular localization, and their role in tumorigenesis. The review also highlights the functional and clinical impact of selective as well as pan kinase inhibitors. Currently, >60 compounds that target AURKs are in preclinical and clinical studies. The drawbacks of existing inhibitors like selectivity, drug resistance and toxicity have also been addressed. Since, majority of inhibitors are Aurora kinase inhibitor (AKI) type-1 that bind to the active (DFGin and Cin) conformation of the kinase, this information may be utilized to design highly selective kinase inhibitors that can be combined with other therapeutic agents for better clinical outcomes.
Collapse
Affiliation(s)
- Deepali Gupta
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Mukesh Kumar
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Sana Saifi
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Shivani Rawat
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - A S Ethayathulla
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India.
| |
Collapse
|
3
|
Jiang K, Bai L, Wang C, Xiao X, Cheng Z, Peng H, Liu S. The Aurora kinase inhibitor AT9283 inhibits Burkitt lymphoma growth by regulating Warburg effect. PeerJ 2023; 11:e16581. [PMID: 38099309 PMCID: PMC10720464 DOI: 10.7717/peerj.16581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/13/2023] [Indexed: 12/17/2023] Open
Abstract
Objective To investigate the effect of the kinase inhibitor AT9283 on Burkitt lymphoma (BL) cells and elucidate the underlying mechanisms. Methods The effect of AT9283 on the proliferation of BL cell lines was tested using the MTT assay. Apoptosis and cell cycle were measured by flow cytometry. The proteins associated with the cell cycle, apoptosis, and the Warburg effect were detected using Western blotting. Alterations in glycolytic metabolism in terms of glucose intake and lactate concentrations were determined by glucose and lactate assays. Results The current study utilized the GEPIA, the Human Protein Atlas (HAP) database and immunohistochemistry to conduct analyses, which revealed a high expression of Aurora kinases and Warburg effect-related proteins in malignant B-cell lymphoma tissues. AT9283 significantly inhibited the cell proliferation of BL cells and induced G2/M arrest. Additionally, AT9283 induced apoptosis in BL cells and reversed the Warburg effect by increasing glucose uptake and reducing lactate production. Moreover, the protein expression of hexokinase 2, pyruvate kinase M2, and lactate dehydrogenase A was significantly suppressed by AT9283, possibly through the inhibition of c-Myc and HIF-1α protein expression. Conclusion The reversal of the Warburg effect in BL cells and the subsequent inhibition of cell proliferation and induction of apoptosis were observed by targeting Aurora A and Aurora B with AT9283. This finding may present new therapeutic options and targets for BL.
Collapse
Affiliation(s)
- Kaiming Jiang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lihong Bai
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Canfei Wang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Xiao
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Zhao Cheng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Sufang Liu
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
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: 8] [Impact Index Per Article: 8.0] [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
|
5
|
Machado CB, DA Silva EL, Dias Nogueira BM, DA Silva JBS, DE Moraes Filho MO, Montenegro RC, DE Moraes MEA, Moreira-Nunes CA. The Relevance of Aurora Kinase Inhibition in Hematological Malignancies. CANCER DIAGNOSIS & PROGNOSIS 2021; 1:111-126. [PMID: 35399305 DOI: 10.21873/cdp.10016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/27/2021] [Indexed: 12/26/2022]
Abstract
Aurora kinases are a family of serine/threonine protein kinases that play a central role in eukaryotic cell division. Overexpression of aurora kinases in cancer and their role as major regulators of the cell cycle quickly inspired the idea that their inhibition might be a potential pathway when treating oncologic patients. Over the past couple of decades, the search for designing and testing of molecules capable of inhibiting aurora activities fueled many pre-clinical and clinical studies. In this study, data from the past 10 years of in vitro and in vivo investigations, as well as clinical trials, utilizing aurora kinase inhibitors as therapeutics for hematological malignancies were compiled and discussed, aiming to highlight potential uses of these inhibitors as a novel monotherapy model or alongside conventional chemotherapies. While there is still much to be elucidated, it is clear that these kinases play a key role in oncogenesis, and their manageable toxicity and potentially synergistic effects still render them a focus of interest for future investigations in combinatorial clinical trials.
Collapse
Affiliation(s)
- Caio Bezerra Machado
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Emerson Lucena DA Silva
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Beatriz Maria Dias Nogueira
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Jean Breno Silveira DA Silva
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Manoel Odorico DE Moraes Filho
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Raquel Carvalho Montenegro
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Caroline Aquino Moreira-Nunes
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
6
|
Serrano-Del Valle A, Reina-Ortiz C, Benedi A, Anel A, Naval J, Marzo I. Future prospects for mitosis-targeted antitumor therapies. Biochem Pharmacol 2021; 190:114655. [PMID: 34129859 DOI: 10.1016/j.bcp.2021.114655] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 11/17/2022]
Abstract
Dysregulation of cell cycle progression is a hallmark of cancer cells. In recent years, efforts have been devoted to the development of new therapies that target proteins involved in cell cycle regulation and mitosis. Novel targeted antimitotic drugs include inhibitors of aurora kinase family, polo-like kinase 1, Mps1, Eg5, CENP-5 and the APC/cyclosome complex. While certain new inhibitors reached the clinical trial stage, most were discontinued due to negative results. However, these therapies should not be readily dismissed. Based on recent advances concerning their mechanisms of action, new strategies could be devised to increase their efficacy and promote further clinical trials. Here we discuss three main lines of action to empower these therapeutic approaches: increasing cell death signals during mitotic arrest, targeting senescent cells and facilitating antitumor immune response through immunogenic cell death (ICD).
Collapse
Affiliation(s)
| | - Chantal Reina-Ortiz
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Andrea Benedi
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Alberto Anel
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Javier Naval
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Isabel Marzo
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain.
| |
Collapse
|
7
|
Abstract
Introduction: Aurora kinases are a family of serine/threonine kinases, and promote mitotic spindle assembly by regulating centrosome duplication and separation. Aurora kinases are overexpressed in a variety of tumor cell lines, thus, the use of Aurora kinase small-molecule inhibitors has become a potential treatment option for cancer.Areas covered: As a continuing review of Aurora kinase inhibitors and their patents published in 2009, 2011 and 2014. Herein, we updated the information for Aurora kinase inhibitors in clinical trials and the patents filed from 2014 to 2020. PubMed, Scopus, SciFinder, and www.clinicaltrials.gov databases were used for searching the clinical information and patents of Aurora kinase inhibitors.Expert opinion: Even though Aurora A or B selective as well as pan inhibitors show preclinical and clinical efficacy, so far, no Aurora kinase inhibitor has been approved for clinical use. Preliminary evidence suggested that highly selective Aurora kinase or multi-target inhibitors as a single agent as well as in combination therapy are still the current main development trend of Aurora kinase inhibitors.
Collapse
Affiliation(s)
- Xue-Li Jing
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Shi-Wu Chen
- School of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
8
|
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: 80] [Impact Index Per Article: 26.7] [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
|
9
|
Antitumor activity of a novel Aurora A/B kinases inhibitor TY-011 against gastric cancer by inducing DNA damage. Anticancer Drugs 2021; 31:440-451. [PMID: 32187025 DOI: 10.1097/cad.0000000000000928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
TY-011, a novel Aurora A/B kinases inhibitor, was found in our previous study to exhibit prominent inhibitory effects on growth of gastric cancer, both in vitro and in vivo. To clarify the mechanisms of TY-011 in inhibiting proliferation of gastric cancer cells, the effects of TY-011 on mitosis, cell cycle, apoptosis and cellular DNA were checked in the present study. Our results showed that TY-011 treatment induced aberrant mitosis, G2/M phase arrest and apoptosis. Importantly, TY-011 induced evident DNA damage in MGC-803 and MKN-45 human gastric cancer cells, which was further characterized as DNA double-strand break. Furthermore, cells treated with TY-011 appeared to generate multiple spindle fibers emanating from several spindle poles, leading to poly-merotelic kinetochore. These results suggested that TY-011 induced abnormal microtubule-kinetochores attachment and thus DNA damage, apoptosis and finally inhibition of cell proliferation of human gastric cancer cells.
Collapse
|
10
|
Du R, Huang C, Liu K, Li X, Dong Z. Targeting AURKA in Cancer: molecular mechanisms and opportunities for Cancer therapy. Mol Cancer 2021; 20:15. [PMID: 33451333 PMCID: PMC7809767 DOI: 10.1186/s12943-020-01305-3] [Citation(s) in RCA: 234] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/29/2020] [Indexed: 12/24/2022] Open
Abstract
Aurora kinase A (AURKA) belongs to the family of serine/threonine kinases, whose activation is necessary for cell division processes via regulation of mitosis. AURKA shows significantly higher expression in cancer tissues than in normal control tissues for multiple tumor types according to the TCGA database. Activation of AURKA has been demonstrated to play an important role in a wide range of cancers, and numerous AURKA substrates have been identified. AURKA-mediated phosphorylation can regulate the functions of AURKA substrates, some of which are mitosis regulators, tumor suppressors or oncogenes. In addition, enrichment of AURKA-interacting proteins with KEGG pathway and GO analysis have demonstrated that these proteins are involved in classic oncogenic pathways. All of this evidence favors the idea of AURKA as a target for cancer therapy, and some small molecules targeting AURKA have been discovered. These AURKA inhibitors (AKIs) have been tested in preclinical studies, and some of them have been subjected to clinical trials as monotherapies or in combination with classic chemotherapy or other targeted therapies.
Collapse
Affiliation(s)
- Ruijuan Du
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China. .,China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, 450008, Henan, China.
| | - Chuntian Huang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.,China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, 450008, Henan, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.,China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, 450008, Henan, China.,The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiang Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China. .,China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, 450008, Henan, China. .,The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, China. .,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China.
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China. .,China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, 450008, Henan, China. .,The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, China. .,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China. .,College of medicine, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
11
|
Induction of differentiation of the acute myeloid leukemia cell line (HL-60) by a securinine dimer. Cell Death Discov 2020; 6:123. [PMID: 33298839 PMCID: PMC7665178 DOI: 10.1038/s41420-020-00354-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/25/2020] [Accepted: 10/07/2020] [Indexed: 01/31/2023] Open
Abstract
Differentiation therapy has been successfully applied clinically in cases of acute promyelocytic leukemia (APL), but few differentiation-induction agents other than all-trans retinoic acid (ATRA) have been discovered clinically. Based on our previously reported neuritogenic differentiation activity of synthetic dimeric derivatives of securinine, we explored the leukemia differentiation-induction activity of such as compound, SN3-L6. It was found that SN3-L6 induces transdifferentiation of both acute myeloid leukemia (AML) and chronic myelogenous leukemia (CML) cells but unexpectedly, a new transdifferentiation pathway from APL cells to morphologically and immunologically normal megakaryocytes and platelets were discovered. SN3-L6 fails to induce transdifferentiation of ATRA–produced mature granulocytes into megakaryocytes, indicating its selectivity between mature and immature cells. SN3-L6 induces CML K562 cells to transdifferentiate into apoptotic megakaryocytes but without platelet formation, indicating a desirable selectivity between different leukemia cells. Our data illuminate a differentiation gap between AML cells and platelets, and promises applications in leukemia differentiation therapy strategy.
Collapse
|
12
|
Torrente L, Maan G, Oumkaltoum Rezig A, Quinn J, Jackson A, Grilli A, Casares L, Zhang Y, Kulesskiy E, Saarela J, Bicciato S, Edwards J, Dinkova-Kostova AT, de la Vega L. High NRF2 Levels Correlate with Poor Prognosis in Colorectal Cancer Patients and with Sensitivity to the Kinase Inhibitor AT9283 In Vitro. Biomolecules 2020; 10:E1365. [PMID: 32992842 PMCID: PMC7600603 DOI: 10.3390/biom10101365] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/15/2020] [Accepted: 09/19/2020] [Indexed: 12/31/2022] Open
Abstract
Aberrant hyperactivation of nuclear factor erythroid 2 (NF-E2) p45-related factor 2 (NRF2) is a common event in many tumour types and associates with resistance to therapy and poor patient prognosis; however, its relevance in colorectal tumours is not well-established. Measuring the expression of surrogate genes for NRF2 activity in silico, in combination with validation in patients' samples, we show that the NRF2 pathway is upregulated in colorectal tumours and that high levels of nuclear NRF2 correlate with a poor patient prognosis. These results highlight the need to overcome the protection provided by NRF2 and present an opportunity to selectively kill cancer cells with hyperactive NRF2. Exploiting the CRISPR/Cas9 technology, we generated colorectal cancer cell lines with hyperactive NRF2 and used them to perform a drug screen. We identified AT9283, an Aurora kinase inhibitor, for its selectivity towards killing cancer cells with hyperactive NRF2 as a consequence to either genetic or pharmacological activation. Our results show that hyperactivation of NRF2 in colorectal cancer cells might present a vulnerability that could potentially be therapeutically exploited by using the Aurora kinase inhibitor AT9283.
Collapse
Affiliation(s)
- Laura Torrente
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK; (L.T.); (G.M.); (A.J.); (L.C.); (Y.Z.); (A.T.D.-K.)
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Gunjit Maan
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK; (L.T.); (G.M.); (A.J.); (L.C.); (Y.Z.); (A.T.D.-K.)
| | - Asma Oumkaltoum Rezig
- Unit of Gastrointestinal Oncology and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK; (A.O.R.); (J.Q.); (J.E.)
| | - Jean Quinn
- Unit of Gastrointestinal Oncology and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK; (A.O.R.); (J.Q.); (J.E.)
| | - Angus Jackson
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK; (L.T.); (G.M.); (A.J.); (L.C.); (Y.Z.); (A.T.D.-K.)
| | - Andrea Grilli
- Department of Life Sciences, University of Modena and Reggio Emilia; via G, Campi 287, 41125 Modena, Italy; (A.G.); (S.B.)
| | - Laura Casares
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK; (L.T.); (G.M.); (A.J.); (L.C.); (Y.Z.); (A.T.D.-K.)
| | - Ying Zhang
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK; (L.T.); (G.M.); (A.J.); (L.C.); (Y.Z.); (A.T.D.-K.)
| | - Evgeny Kulesskiy
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Tukholmankatu 8, FI-00290 Helsinki, Finland; (E.K.); (J.S.)
| | - Jani Saarela
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Tukholmankatu 8, FI-00290 Helsinki, Finland; (E.K.); (J.S.)
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia; via G, Campi 287, 41125 Modena, Italy; (A.G.); (S.B.)
| | - Joanne Edwards
- Unit of Gastrointestinal Oncology and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK; (A.O.R.); (J.Q.); (J.E.)
| | - Albena T. Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK; (L.T.); (G.M.); (A.J.); (L.C.); (Y.Z.); (A.T.D.-K.)
- Departments of Medicine and Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Laureano de la Vega
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK; (L.T.); (G.M.); (A.J.); (L.C.); (Y.Z.); (A.T.D.-K.)
| |
Collapse
|
13
|
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: 30] [Impact Index Per Article: 6.0] [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
|
14
|
Mills CC, Kolb EA, Sampson VB. Recent Advances of Cell-Cycle Inhibitor Therapies for Pediatric Cancer. Cancer Res 2017; 77:6489-6498. [PMID: 29097609 DOI: 10.1158/0008-5472.can-17-2066] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/24/2017] [Accepted: 09/19/2017] [Indexed: 12/21/2022]
Abstract
This review describes the pivotal roles of cell-cycle and checkpoint regulators and discusses development of specific cell-cycle inhibitors for therapeutic use for pediatric cancer. The mechanism of action as well as the safety and tolerability of drugs in pediatric patients, including compounds that target CDK4/CDK6 (palbociclib, ribociclib, and abemaciclib), aurora kinases (AT9283 and MLN8237), Wee1 kinase (MK-1775), KSP (ispinesib), and tubulin (taxanes, vinca alkaloids), are presented. The design of mechanism-based combinations that exploit the cross-talk of signals activated by cell-cycle arrest, as well as pediatric-focused drug development, are critical for the advancement of drugs for rare childhood diseases. Cancer Res; 77(23); 6489-98. ©2017 AACR.
Collapse
Affiliation(s)
| | - E A Kolb
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Valerie B Sampson
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware.
| |
Collapse
|
15
|
Duong JK, Griffin MJ, Hargrave D, Vormoor J, Edwards D, Boddy AV. A population pharmacokinetic model of AT9283 in adults and children to predict the maximum tolerated dose in children with leukaemia. Br J Clin Pharmacol 2017; 83:1713-1722. [PMID: 28177130 PMCID: PMC5510070 DOI: 10.1111/bcp.13260] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/19/2017] [Accepted: 02/06/2017] [Indexed: 01/23/2023] Open
Abstract
AIMS AT9283 is used to treat patients with solid tumours and patients with leukaemia. However, the maximum tolerated dose (MTD) for children with leukaemia remains unknown due to early termination of the Phase I trial. The aim of this study was to develop a population model of AT9283 to describe the pharmacokinetics in adults and children and to estimate the MTD in children with leukaemia. METHODS Data from Phase I dose-escalation studies in adults and children were used to build a population pharmacokinetic model (NONMEM v7.3). Potential covariates investigated included body weight, body surface area (BSA), glomerular filtration rate (GFR), age and sex. Model-derived area under the concentration-time curve was used to investigate the relationship between dose and exposure in adults and children. RESULTS The plasma concentrations of AT9283 (n = 1770) from 92 patients (53 adults, 39 children) were used to build a two-compartment model with all pharmacokinetic parameters scaled using body weight. Renal function (GFR), but not BSA, was a significant covariate for the clearance of AT9283. In children with leukaemia (median weight 16 kg), a flat dose of 500 mg 72 h-1 provided similar drug exposures at the MTD as the adult population. The estimated MTD for children with leukaemia, therefore, is 30 mg kg-1 72 h-1 . CONCLUSION For adults, GFR was a significant predictor of clearance, whilst body-weight based dosing was more useful than BSA in determining the drug exposure in children. The MTD was estimated to be 30 mg kg-1 72 h-1 children with leukaemia.
Collapse
Affiliation(s)
- Janna K. Duong
- Faculty of PharmacyThe University of SydneySydneyAustralia
| | | | - Darren Hargrave
- Great Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Josef Vormoor
- Northern Institute for Cancer ResearchNewcastle upon TyneUK
- Great North Children's HospitalNewcastle upon TyneUK
| | | | - Alan V. Boddy
- Faculty of PharmacyThe University of SydneySydneyAustralia
| |
Collapse
|
16
|
Zwaan CM, Den Boer ML. The unpredictable Northern light: The rise and fall of the aurora kinase inhibitors? Pediatr Blood Cancer 2017; 64. [PMID: 28205329 DOI: 10.1002/pbc.26418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 11/30/2016] [Indexed: 11/10/2022]
Affiliation(s)
- C M Zwaan
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - M L Den Boer
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|