1
|
Cavalu S, Abdelhamid AM, Saber S, Elmorsy EA, Hamad RS, Abdel-Reheim MA, Yahya G, Salama MM. Cell cycle machinery in oncology: A comprehensive review of therapeutic targets. FASEB J 2024; 38:e23734. [PMID: 38847486 DOI: 10.1096/fj.202400769r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/20/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024]
Abstract
The cell cycle is tightly regulated to ensure controlled cell proliferation. Dysregulation of the cell cycle machinery is a hallmark of cancer that leads to unchecked growth. This review comprehensively analyzes key molecular regulators of the cell cycle and how they contribute to carcinogenesis when mutated or overexpressed. It focuses on cyclins, cyclin-dependent kinases (CDKs), CDK inhibitors, checkpoint kinases, and mitotic regulators as therapeutic targets. Promising strategies include CDK4/6 inhibitors like palbociclib, ribociclib, and abemaciclib for breast cancer treatment. Other possible targets include the anaphase-promoting complex/cyclosome (APC/C), Skp2, p21, and aurora kinase inhibitors. However, challenges with resistance have limited clinical successes so far. Future efforts should focus on combinatorial therapies, next-generation inhibitors, and biomarkers for patient selection. Targeting the cell cycle holds promise but further optimization is necessary to fully exploit it as an anti-cancer strategy across diverse malignancies.
Collapse
Affiliation(s)
- Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Amir Mohamed Abdelhamid
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Elsayed A Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah, Saudi Arabia
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
- Central Laboratory, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, Egypt
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Al Sharqia, Egypt
| | - Mohamed M Salama
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| |
Collapse
|
2
|
Zarrabi A, Perrin D, Kavoosi M, Sommer M, Sezen S, Mehrbod P, Bhushan B, Machaj F, Rosik J, Kawalec P, Afifi S, Bolandi SM, Koleini P, Taheri M, Madrakian T, Łos MJ, Lindsey B, Cakir N, Zarepour A, Hushmandi K, Fallah A, Koc B, Khosravi A, Ahmadi M, Logue S, Orive G, Pecic S, Gordon JW, Ghavami S. Rhabdomyosarcoma: Current Therapy, Challenges, and Future Approaches to Treatment Strategies. Cancers (Basel) 2023; 15:5269. [PMID: 37958442 PMCID: PMC10650215 DOI: 10.3390/cancers15215269] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/18/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Rhabdomyosarcoma is a rare cancer arising in skeletal muscle that typically impacts children and young adults. It is a worldwide challenge in child health as treatment outcomes for metastatic and recurrent disease still pose a major concern for both basic and clinical scientists. The treatment strategies for rhabdomyosarcoma include multi-agent chemotherapies after surgical resection with or without ionization radiotherapy. In this comprehensive review, we first provide a detailed clinical understanding of rhabdomyosarcoma including its classification and subtypes, diagnosis, and treatment strategies. Later, we focus on chemotherapy strategies for this childhood sarcoma and discuss the impact of three mechanisms that are involved in the chemotherapy response including apoptosis, macro-autophagy, and the unfolded protein response. Finally, we discuss in vivo mouse and zebrafish models and in vitro three-dimensional bioengineering models of rhabdomyosarcoma to screen future therapeutic approaches and promote muscle regeneration.
Collapse
Affiliation(s)
- Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Türkiye; (A.Z.); (A.Z.)
| | - David Perrin
- Section of Orthopaedic Surgery, Department of Surgery, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; (D.P.); (M.S.)
| | - Mahboubeh Kavoosi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Biotechnology Center, Silesian University of Technology, 8 Krzywousty St., 44-100 Gliwice, Poland;
| | - Micah Sommer
- Section of Orthopaedic Surgery, Department of Surgery, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; (D.P.); (M.S.)
- Section of Physical Medicine and Rehabilitation, Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Serap Sezen
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Türkiye; (S.S.); (N.C.); (B.K.)
| | - Parvaneh Mehrbod
- Department of Influenza and Respiratory Viruses, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Bhavya Bhushan
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Science, McGill University, Montreal, QC H3A 0C7, Canada
| | - Filip Machaj
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Jakub Rosik
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Philip Kawalec
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Section of Neurosurgery, Department of Surgery, University of Manitoba, Health Sciences Centre, Winnipeg, MB R3A 1R9, Canada
| | - Saba Afifi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Seyed Mohammadreza Bolandi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Peiman Koleini
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Mohsen Taheri
- Genetics of Non-Communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran;
| | - Tayyebeh Madrakian
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (T.M.); (M.A.)
| | - Marek J. Łos
- Biotechnology Center, Silesian University of Technology, 8 Krzywousty St., 44-100 Gliwice, Poland;
| | - Benjamin Lindsey
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Nilufer Cakir
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Türkiye; (S.S.); (N.C.); (B.K.)
| | - Atefeh Zarepour
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Türkiye; (A.Z.); (A.Z.)
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran 1419963114, Iran;
| | - Ali Fallah
- Integrated Manufacturing Technologies Research and Application Center, Sabanci University, Tuzla, Istanbul 34956, Türkiye;
| | - Bahattin Koc
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Türkiye; (S.S.); (N.C.); (B.K.)
- Integrated Manufacturing Technologies Research and Application Center, Sabanci University, Tuzla, Istanbul 34956, Türkiye;
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Türkiye
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Türkiye;
| | - Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (T.M.); (M.A.)
| | - Susan Logue
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), 01007 Vitoria-Gasteiz, Spain;
- University Institute for Regenerative Medicine and Oral Implantology–UIRMI (UPV/EHU-Fundación Eduardo Anitua), 01007 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, 01006 Vitoria-Gasteiz, Spain
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, USA;
| | - Joseph W. Gordon
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- College of Nursing, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555 Katowice, Poland
- Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| |
Collapse
|
3
|
Shah K, Nasimian A, Ahmed M, Al Ashiri L, Denison L, Sime W, Bendak K, Kolosenko I, Siino V, Levander F, Palm-Apergi C, Massoumi R, Lock RB, Kazi JU. PLK1 as a cooperating partner for BCL2-mediated antiapoptotic program in leukemia. Blood Cancer J 2023; 13:139. [PMID: 37679323 PMCID: PMC10484999 DOI: 10.1038/s41408-023-00914-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/15/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
The deregulation of BCL2 family proteins plays a crucial role in leukemia development. Therefore, pharmacological inhibition of this family of proteins is becoming a prevalent treatment method. However, due to the emergence of primary and acquired resistance, efficacy is compromised in clinical or preclinical settings. We developed a drug sensitivity prediction model utilizing a deep tabular learning algorithm for the assessment of venetoclax sensitivity in T-cell acute lymphoblastic leukemia (T-ALL) patient samples. Through analysis of predicted venetoclax-sensitive and resistant samples, PLK1 was identified as a cooperating partner for the BCL2-mediated antiapoptotic program. This finding was substantiated by additional data obtained through phosphoproteomics and high-throughput kinase screening. Concurrent treatment using venetoclax with PLK1-specific inhibitors and PLK1 knockdown demonstrated a greater therapeutic effect on T-ALL cell lines, patient-derived xenografts, and engrafted mice compared with using each treatment separately. Mechanistically, the attenuation of PLK1 enhanced BCL2 inhibitor sensitivity through upregulation of BCL2L13 and PMAIP1 expression. Collectively, these findings underscore the dependency of T-ALL on PLK1 and postulate a plausible regulatory mechanism.
Collapse
Affiliation(s)
- Kinjal Shah
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ahmad Nasimian
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Mehreen Ahmed
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Lina Al Ashiri
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Linn Denison
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Wondossen Sime
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Katerina Bendak
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Iryna Kolosenko
- Department of Laboratory Medicine, Biomolecular & Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Valentina Siino
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Fredrik Levander
- Department of Immunotechnology, Lund University, Lund, Sweden
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Lund University, Lund, Sweden
| | - Caroline Palm-Apergi
- Department of Laboratory Medicine, Biomolecular & Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ramin Massoumi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Julhash U Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden.
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
4
|
Reda M, Ngamcherdtrakul W, Nelson MA, Siriwon N, Wang R, Zaidan HY, Bejan DS, Reda S, Hoang NH, Crumrine NA, Rehwaldt JPC, Bindal A, Mills GB, Gray JW, Yantasee W. Development of a nanoparticle-based immunotherapy targeting PD-L1 and PLK1 for lung cancer treatment. Nat Commun 2022; 13:4261. [PMID: 35871223 PMCID: PMC9308817 DOI: 10.1038/s41467-022-31926-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 07/11/2022] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting PD-L1 and PD-1 have improved survival in a subset of patients with advanced non-small cell lung cancer (NSCLC). However, only a minority of NSCLC patients respond to ICIs, highlighting the need for superior immunotherapy. Herein, we report on a nanoparticle-based immunotherapy termed ARAC (Antigen Release Agent and Checkpoint Inhibitor) designed to enhance the efficacy of PD-L1 inhibitor. ARAC is a nanoparticle co-delivering PLK1 inhibitor (volasertib) and PD-L1 antibody. PLK1 is a key mitotic kinase that is overexpressed in various cancers including NSCLC and drives cancer growth. Inhibition of PLK1 selectively kills cancer cells and upregulates PD-L1 expression in surviving cancer cells thereby providing opportunity for ARAC targeted delivery in a feedforward manner. ARAC reduces effective doses of volasertib and PD-L1 antibody by 5-fold in a metastatic lung tumor model (LLC-JSP) and the effect is mainly mediated by CD8+ T cells. ARAC also shows efficacy in another lung tumor model (KLN-205), which does not respond to CTLA-4 and PD-1 inhibitor combination. This study highlights a rational combination strategy to augment existing therapies by utilizing our nanoparticle platform that can load multiple cargo types at once. Only a minority of patients with non-small cell lung cancer (NSCLC) respond to immune checkpoint inhibitors. Here the authors design a nanosystem for the co-delivery of a PLK1 inhibitor and PD-L1 antibody, showing anti-tumor immune responses in preclinical lung cancer models.
Collapse
|
5
|
Kuburich NA, den Hollander P, Pietz JT, Mani SA. Vimentin and cytokeratin: Good alone, bad together. Semin Cancer Biol 2022; 86:816-826. [PMID: 34953942 PMCID: PMC9213573 DOI: 10.1016/j.semcancer.2021.12.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/26/2021] [Accepted: 12/14/2021] [Indexed: 01/27/2023]
Abstract
The cytoskeleton plays an integral role in maintaining the integrity of epithelial cells. Epithelial cells primarily employ cytokeratin in their cytoskeleton, whereas mesenchymal cells use vimentin. During the epithelial-mesenchymal transition (EMT), cytokeratin-positive epithelial cells begin to express vimentin. EMT induces stem cell properties and drives metastasis, chemoresistance, and tumor relapse. Most studies of the functions of cytokeratin and vimentin have relied on the use of either epithelial or mesenchymal cell types. However, it is important to understand how these two cytoskeleton intermediate filaments function when co-expressed in cells undergoing EMT. Here, we discuss the individual and shared functions of cytokeratin and vimentin that coalesce during EMT and how alterations in intermediate filament expression influence carcinoma progression.
Collapse
Affiliation(s)
- Nick A Kuburich
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Petra den Hollander
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Jordan T Pietz
- Department of Creative Services, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States.
| |
Collapse
|
6
|
Bhujbal SP, Kim H, Bae H, Hah JM. Design and Synthesis of Aminopyrimidinyl Pyrazole Analogs as PLK1 Inhibitors Using Hybrid 3D-QSAR and Molecular Docking. Pharmaceuticals (Basel) 2022; 15:1170. [PMID: 36297281 PMCID: PMC9610081 DOI: 10.3390/ph15101170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/02/2022] [Accepted: 09/16/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer continues to be one of the world's most severe public health issues. Polo-like kinase 1 (PLK1) is one of the most studied members of the polo-like kinase subfamily of serine/threonine protein kinases. PLK1 is a key mitotic regulator responsible for cell cycle processes, such as mitosis initiation, bipolar mitotic spindle formation, centrosome maturation, the metaphase to anaphase transition, and mitotic exit, whose overexpression is often associated with oncogenesis. Moreover, it is also involved in DNA damage response, autophagy, cytokine signaling, and apoptosis. Due to its fundamental role in cell cycle regulation, PLK1 has been linked to various types of cancer onset and progression, such as lung, colon, prostate, ovary, breast cancer, melanoma, and AML. Hence, PLK1 is recognized as a critical therapeutic target in the treatment of various proliferative diseases. PLK1 inhibitors developed in recent years have been researched and studied through clinical trials; however, most of them have failed because of their toxicity and poor therapeutic response. To design more potent and selective PLK1 inhibitors, we performed a receptor-based hybrid 3D-QSAR study of two datasets, possessing similar common scaffolds. The developed hybrid CoMFA (q2 = 0.628, r2 = 0.905) and CoMSIA (q2 = 0.580, r2 = 0.895) models showed admissible statistical results. Comprehensive, molecular docking of one of the most active compounds from the dataset and hybrid 3D-QSAR studies revealed important active site residues of PLK1 and requisite structural characteristics of ligand to design potent PLK1 inhibitors. Based on this information, we have proposed approximately 38 PLK1 inhibitors. The newly designed PLK1 inhibitors showed higher activity (predicted pIC50) than the most active compounds of all the derivatives selected for this study. We selected and synthesized two compounds, which were ultimately found to possess good IC50 values. Our design strategy provides insight into development of potent and selective PLK1 inhibitors.
Collapse
Affiliation(s)
- Swapnil P. Bhujbal
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 426-791, Korea
- Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 426-791, Korea
| | - Hyejin Kim
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 426-791, Korea
- Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 426-791, Korea
| | - Hyunah Bae
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 426-791, Korea
- Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 426-791, Korea
| | - Jung-Mi Hah
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 426-791, Korea
- Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 426-791, Korea
| |
Collapse
|
7
|
Chiappa M, Petrella S, Damia G, Broggini M, Guffanti F, Ricci F. Present and Future Perspective on PLK1 Inhibition in Cancer Treatment. Front Oncol 2022; 12:903016. [PMID: 35719948 PMCID: PMC9201472 DOI: 10.3389/fonc.2022.903016] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022] Open
Abstract
Polo-like kinase 1 (PLK1) is the principle member of the well conserved serine/threonine kinase family. PLK1 has a key role in the progression of mitosis and recent evidence suggest its important involvement in regulating the G2/M checkpoint, in DNA damage and replication stress response, and in cell death pathways. PLK1 expression is tightly spatially and temporally regulated to ensure its nuclear activation at the late S-phase, until the peak of expression at the G2/M-phase. Recently, new roles of PLK1 have been reported in literature on its implication in the regulation of inflammation and immunological responses. All these biological processes are altered in tumors and, considering that PLK1 is often found overexpressed in several tumor types, its targeting has emerged as a promising anti-cancer therapeutic strategy. In this review, we will summarize the evidence suggesting the role of PLK1 in response to DNA damage, including DNA repair, cell cycle progression, epithelial to mesenchymal transition, cell death pathways and cancer-related immunity. An update of PLK1 inhibitors currently investigated in preclinical and clinical studies, in monotherapy and in combination with existing chemotherapeutic drugs and targeted therapies will be discussed.
Collapse
Affiliation(s)
- Michela Chiappa
- Laboratory of Experimental Oncology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Serena Petrella
- Laboratory of Experimental Oncology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Giovanna Damia
- Laboratory of Experimental Oncology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Massimo Broggini
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Federica Guffanti
- Laboratory of Experimental Oncology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Francesca Ricci
- Laboratory of Experimental Oncology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| |
Collapse
|
8
|
Platzbecker U, Chromik J, Krönke J, Handa H, Strickland S, Miyazaki Y, Wermke M, Sakamoto W, Tachibana Y, Taube T, Germing U. Volasertib as a monotherapy or in combination with azacitidine in patients with myelodysplastic syndrome, chronic myelomonocytic leukemia, or acute myeloid leukemia: summary of three phase I studies. BMC Cancer 2022; 22:569. [PMID: 35597904 PMCID: PMC9124414 DOI: 10.1186/s12885-022-09622-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This report summarizes three phase I studies evaluating volasertib, a polo-like kinase inhibitor, plus azacitidine in adults with myelodysplastic syndromes (MDS), chronic myelomonocytic leukemia, or acute myeloid leukemia. METHODS Patients received intravenous volasertib in 28-day cycles (dose-escalation schedules). In Part 1 of 1230.33 (Study 1; NCT01957644), patients received 250-350 mg volasertib on day (D)1 and D15; in Part 2, patients received different schedules [A, D1: 170 mg/m2; B, D7: 170 mg/m2; C, D1 and D7: 110 mg/m2]. In 1230.35 (Study 2; NCT02201329), patients received 200-300 mg volasertib on D1 and D15. In 1230.43 (Study 3; NCT02721875), patients received 110 mg/m2 volasertib on D1 and D8. All patients in Studies 1 and 2, and approximately half of the patients in Study 3, were scheduled to receive subcutaneous azacitidine 75 mg/m2 on D1-7. RESULTS Overall, 22 patients were treated (17 with MDS; 12 previously untreated). Across Studies 1 and 2 (n = 21), the most common drug-related adverse events were hematological (thrombocytopenia [n = 11]; neutropenia [n = 8]). All dose-limiting toxicities were grade 4 thrombocytopenia. The only treated patient in Study 3 experienced 18 adverse events following volasertib monotherapy. Studies 1 and 2 showed preliminary activity (objective response rates: 25 and 40%). CONCLUSIONS The safety of volasertib with azacitidine in patients with MDS was consistent with other volasertib studies. All studies were terminated prematurely following the discontinuation of volasertib for non-clinical reasons by Boehringer Ingelheim; however, safety information on volasertib plus azacitidine are of interest for future studies in other diseases.
Collapse
Affiliation(s)
- Uwe Platzbecker
- Medical Clinic and Policlinic I, Hematology and Cellular Therapy, University Hospital Leipzig, Johannisallee 32, D-04103, Leipzig, Germany.
| | - Joerg Chromik
- Department of Hematology and Medical Oncology, University Hospital Frankfurt, Frankfurt, Germany
| | - Jan Krönke
- Department of Internal Medicine, University Hospital of Ulm, Ulm, Germany
| | - Hiroshi Handa
- Department of Hematology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Stephen Strickland
- Division of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Yasushi Miyazaki
- Department of Hematology, Nagasaki University Hospital, Nagasaki City, Japan
| | - Martin Wermke
- NCT/UCC Early Clinical Trial Unit, Technical University Dresden, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Wataru Sakamoto
- Biostatistics and Data Science Japan, Medical Division, Nippon Boehringer Ingelheim, Tokyo, Japan
| | | | - Tillmann Taube
- Therapeutic Area Oncology Medicine, Boehringer Ingelheim International, Biberach, Germany
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine University, Dusseldorf, Germany
| |
Collapse
|
9
|
Design and Synthesis of a Novel 4-aryl-N-(2-alkoxythieno [2,3-b]pyrazine-3-yl)-4-arylpiperazine-1-carboxamide DGG200064 Showed Therapeutic Effect on Colon Cancer through G2/M Arrest. Pharmaceuticals (Basel) 2022; 15:ph15050502. [PMID: 35631329 PMCID: PMC9143821 DOI: 10.3390/ph15050502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 02/04/2023] Open
Abstract
Cancer cells are characterized by an abnormal cell cycle. Therefore, the cell cycle has been a potential target for cancer therapeutic agents. We developed a new lead compound, DGG200064 (7c) with a 2-alkoxythieno [2,3-b]pyrazine-3-yl)-4-arylpiperazine-1-carboxamide core skeleton. To evaluate its properties, compound DGG200064 was tested in vivo through a xenograft mouse model of colorectal cancer using HCT116 cells. The in vivo results showed high cell growth inhibition efficacy. Our results confirmed that the newly synthesized DGG200064 inhibits the growth of colorectal cancer cells by inducing G2/M arrest. Unlike the known cell cycle inhibitors, DGG200064 (GI50 = 12 nM in an HCT116 cell-based assay) induced G2/M arrest by selectively inhibiting the interaction of FBXW7 and c-Jun proteins. Additionally, the physicochemical properties of the lead compounds were analyzed. Based on the results of the study, we suggested further development of DGG200064 as a novel oral anti-colorectal cancer drug.
Collapse
|
10
|
Tontsch-Grunt U, Traexler PE, Baum A, Musa H, Marzin K, Wang S, Trapani F, Engelhardt H, Solca F. Therapeutic impact of BET inhibitor BI 894999 treatment: backtranslation from the clinic. Br J Cancer 2022; 127:577-586. [PMID: 35444289 PMCID: PMC9346113 DOI: 10.1038/s41416-022-01815-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/24/2022] [Accepted: 03/31/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND BET inhibitors have been tested in several clinical trials where, despite encouraging preclinical results, substantial clinical benefit in monotherapy remains limited. This work illustrates the translational challenges and reports new data around the novel BET inhibitor, BI 894999. At clinically achievable concentrations, mechanistic studies were carried out to study pathway modulation and rational drug combinations. METHODS BRD-NUT fusions are oncogenic drivers in NUT carcinoma (NC). The effects of BI 894999 on proliferation, chromatin binding and pathway modulation were studied in NC in vitro. These studies were complemented by efficacy studies either as a single agent or in combination with the clinical p300/CBP inhibitor CCS1477. RESULTS Based on the modelling of preclinical and clinical data, we proposed and implemented a new clinical scheduling regimen. This led to plasma levels sufficient to fully dislodge BRD-NUT from chromatin and to sustained and pronounced pharmacodynamic (PD) modulation of HEXIM1 and HIST2H2BF. Platelet counts in patient blood samples were improved compared to previous schedules. Rational combination studies of BI 894999 performed at clinically meaningful concentrations led to tumour regressions in all NC xenograft models tested. CONCLUSIONS BI 894999 holds significant potential as a combination drug and CCS1477 p300/CBP inhibitor is a promising partner for future clinical trials.
Collapse
Affiliation(s)
| | | | - Anke Baum
- Boehringer Ingelheim RCV GmbH & Co KG, A-1120, Vienna, Austria
| | - Hanny Musa
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Kristell Marzin
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Shaonan Wang
- Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim am Rhein, Germany
| | | | | | - Flavio Solca
- Boehringer Ingelheim RCV GmbH & Co KG, A-1120, Vienna, Austria
| |
Collapse
|
11
|
Bewersdorf JP, Zeidan AM. Polo-like kinase inhibition as a therapeutic target in acute myeloid leukemia. Oncotarget 2021; 12:1314-1317. [PMID: 34194628 PMCID: PMC8238245 DOI: 10.18632/oncotarget.27919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Affiliation(s)
- Jan Philipp Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA.,Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA
| |
Collapse
|
12
|
Carr RM, Vorobyev D, Lasho T, Marks DL, Tolosa EJ, Vedder A, Almada LL, Yurcheko A, Padioleau I, Alver B, Coltro G, Binder M, Safgren SL, Horn I, You X, Solary E, Balasis ME, Berger K, Hiebert J, Witzig T, Buradkar A, Graf T, Valent P, Mangaonkar AA, Robertson KD, Howard MT, Kaufmann SH, Pin C, Fernandez-Zapico ME, Geissler K, Droin N, Padron E, Zhang J, Nikolaev S, Patnaik MM. RAS mutations drive proliferative chronic myelomonocytic leukemia via a KMT2A-PLK1 axis. Nat Commun 2021; 12:2901. [PMID: 34006870 PMCID: PMC8131698 DOI: 10.1038/s41467-021-23186-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/20/2021] [Indexed: 12/24/2022] Open
Abstract
Proliferative chronic myelomonocytic leukemia (pCMML), an aggressive CMML subtype, is associated with dismal outcomes. RAS pathway mutations, mainly NRASG12D, define the pCMML phenotype as demonstrated by our exome sequencing, progenitor colony assays and a Vav-Cre-NrasG12D mouse model. Further, these mutations promote CMML transformation to acute myeloid leukemia. Using a multiomics platform and biochemical and molecular studies we show that in pCMML RAS pathway mutations are associated with a unique gene expression profile enriched in mitotic kinases such as polo-like kinase 1 (PLK1). PLK1 transcript levels are shown to be regulated by an unmutated lysine methyl-transferase (KMT2A) resulting in increased promoter monomethylation of lysine 4 of histone 3. Pharmacologic inhibition of PLK1 in RAS mutant patient-derived xenografts, demonstrates the utility of personalized biomarker-driven therapeutics in pCMML.
Collapse
MESH Headings
- Animals
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- GTP Phosphohydrolases/genetics
- GTP Phosphohydrolases/metabolism
- Gene Expression Profiling/methods
- Gene Expression Regulation, Leukemic
- Histone-Lysine N-Methyltransferase/genetics
- Histone-Lysine N-Methyltransferase/metabolism
- Kaplan-Meier Estimate
- Leukemia, Myelomonocytic, Chronic/genetics
- Leukemia, Myelomonocytic, Chronic/metabolism
- Leukemia, Myelomonocytic, Chronic/therapy
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Mutation
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Signal Transduction/genetics
- Stem Cell Transplantation/methods
- Transplantation, Homologous
- Exome Sequencing/methods
- Xenograft Model Antitumor Assays/methods
- Polo-Like Kinase 1
- Mice
Collapse
Affiliation(s)
- Ryan M Carr
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, MN, USA
| | - Denis Vorobyev
- INSERM U981, Gustave Roussy Cancer Center, Villejuif, France
| | - Terra Lasho
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
| | - David L Marks
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, MN, USA
| | - Ezequiel J Tolosa
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, MN, USA
| | - Alexis Vedder
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center, Tampa, FL, USA
| | - Luciana L Almada
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, MN, USA
| | - Andrey Yurcheko
- INSERM U981, Gustave Roussy Cancer Center, Villejuif, France
| | | | - Bonnie Alver
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, MN, USA
| | - Giacomo Coltro
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
| | - Moritz Binder
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
| | - Stephanie L Safgren
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, MN, USA
| | - Isaac Horn
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, MN, USA
| | - Xiaona You
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Eric Solary
- INSERM U1170 and Department of Hematology, Gustave Roussy Cancer Center, Villejuif, France
| | - Maria E Balasis
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center, Tampa, FL, USA
| | - Kurt Berger
- London Regional Transgenic and Gene Targeting Facility, Lawson Health Research Institute University of Western Ontario, London, ON, Canada
| | - James Hiebert
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
| | - Thomas Witzig
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
| | - Ajinkya Buradkar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
| | - Temeida Graf
- 5TH Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Peter Valent
- 5TH Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | | | - Keith D Robertson
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, MN, USA
| | - Matthew T Howard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, MN, USA
| | - Scott H Kaufmann
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
| | - Christopher Pin
- London Regional Transgenic and Gene Targeting Facility, Lawson Health Research Institute University of Western Ontario, London, ON, Canada
| | | | | | - Nathalie Droin
- INSERM U1170 and Department of Hematology, Gustave Roussy Cancer Center, Villejuif, France
| | - Eric Padron
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center, Tampa, FL, USA
| | - Jing Zhang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Sergey Nikolaev
- INSERM U981, Gustave Roussy Cancer Center, Villejuif, France.
| | - Mrinal M Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA.
| |
Collapse
|
13
|
Kolosenko I, Goroshchuk O, Vidarsdottir L, Björklund AC, Dowdy SF, Palm-Apergi C. RNAi prodrugs decrease elevated mRNA levels of Polo-like kinase 1 in ex vivo cultured primary cells from pediatric acute myeloid leukemia patients. FASEB J 2021; 35:e21476. [PMID: 33788972 DOI: 10.1096/fj.202002454rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/20/2021] [Accepted: 02/11/2021] [Indexed: 12/19/2022]
Abstract
Polo-like kinase 1 (Plk1) is an important regulator of the cell cycle and it is frequently overexpressed in cancer cells. Several small molecule inhibitors have been developed to target Plk1 and some of them have reached clinical trials in adults with acute myeloid leukemia (AML). Pediatric AML patients have a poor prognosis and survivors suffer from long-term side effects. As adult AML cells have an elevated expression of Plk1, AML is a disease candidate for Plk1 inhibition. However, the relative success of clinical trials have been hampered by adverse reactions. Herein, PLK1-targeting RNA interference (RNAi) prodrugs that enter cells without a transfection reagent are used to target PLK1 selectively in primary cells from pediatric AML patients. We show that PLK1 and PLK4 mRNA expression are significantly higher in pediatric AML patients when compared to healthy donors and that PLK1 is downregulated by on average 50% using RNAi prodrugs without a significant effect on other PLK family members. In addition, the RNAi prodrug-induced decrease in PLK1 can be used to potentiate the effect of cytarabine. In summary, PLK1-targeting RNAi prodrugs can decrease the elevated levels of PLK1 in primary cells from pediatric AML patients and sensitize pediatric AML cells to chemotherapeutics.
Collapse
MESH Headings
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Case-Control Studies
- Cell Cycle
- Cell Cycle Proteins/antagonists & inhibitors
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Movement
- Cell Proliferation
- Child
- Gene Expression Regulation, Neoplastic
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Prodrugs/administration & dosage
- Prognosis
- Protein Kinase Inhibitors/pharmacology
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- RNA Interference
- RNA, Messenger/antagonists & inhibitors
- RNA, Messenger/genetics
- Tumor Cells, Cultured
- Tumor Suppressor Proteins/antagonists & inhibitors
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- Polo-Like Kinase 1
Collapse
Affiliation(s)
- Iryna Kolosenko
- Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Oksana Goroshchuk
- Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Linda Vidarsdottir
- Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ann-Charlotte Björklund
- Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Steven F Dowdy
- Department of Cellular & Molecular Medicine, UCSD School of Medicine, La Jolla, CA, USA
| | - Caroline Palm-Apergi
- Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
14
|
Lin SF, Yeh CN, Huang YT, Chou TC, Wong RJ. Therapeutic inhibition of polo-like kinases in anaplastic thyroid cancer. Cancer Sci 2021; 112:803-814. [PMID: 33306266 PMCID: PMC7893987 DOI: 10.1111/cas.14769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 12/16/2022] Open
Abstract
Polo‐like kinases (PLKs) are potent regulators of cell proliferation and cell survival. Polo‐like kinases are potential targets in the treatment of anaplastic thyroid cancer (ATC), a rare but deadly disease. The therapeutic effects of volasertib, a PLK inhibitor, was evaluated for the treatment of ATC either alone or in combination with sorafenib. Volasertib decreased cell viability in three ATC cell lines (8505C, 8305C, and KAT18) in a dose‐dependent manner. Volasertib caused ATC cells to accumulate in G2/M phase, activated caspase‐3 activity, and induced apoptosis. Combination therapy using volasertib and sorafenib in ATC cells showed mostly synergistic effects. In vivo studies revealed that combination therapy of volasertib and sorafenib was effective in the treatment of 8505C xenografts. Single‐agent volasertib treatment was sufficient to retard 8305C tumor growth. No substantial morbidity was observed in animals that received either single‐agent or combination treatment. These preclinical findings suggest that volasertib could be an effective drug in treating ATC.
Collapse
Affiliation(s)
- Shu-Fu Lin
- Department of Internal Medicine, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan.,Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Nan Yeh
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yu-Tung Huang
- Center for Big Data Analytics and Statistics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ting-Chao Chou
- Laboratory of Preclinical Pharmacology Core, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Richard J Wong
- Head and Neck Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
15
|
Cortes J, Podoltsev N, Kantarjian H, Borthakur G, Zeidan AM, Stahl M, Taube T, Fagan N, Rajeswari S, Uy GL. Phase 1 dose escalation trial of volasertib in combination with decitabine in patients with acute myeloid leukemia. Int J Hematol 2021; 113:92-99. [PMID: 32951163 DOI: 10.1007/s12185-020-02994-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/19/2020] [Accepted: 09/07/2020] [Indexed: 11/30/2022]
Abstract
Polo-like kinase 1 (PLK1) regulates mitotic checkpoints and cell division. PLK1 overexpression is reported in numerous cancers, including acute myeloid leukemia (AML), and is associated with poor prognosis. Volasertib is a selective, potent cell-cycle kinase inhibitor that targets PLK to induce mitotic arrest and apoptosis. This phase 1 trial investigated the maximum tolerated dose (MTD), safety, pharmacokinetics, and anti-leukemic activity of volasertib in combination with decitabine in AML patients aged ≥ 65 years. Thirteen patients were treated with escalating volasertib doses (3 + 3 design; 300 mg, 350 mg, and 400 mg) plus standard-dose decitabine. Dose-limiting toxicity was reported in one patient in cycle 1; the MTD of volasertib in combination with decitabine was determined as 400 mg. The most common treatment-emergent adverse events were febrile neutropenia, pneumonia, and decreased appetite. Objective response rate was 23%. The combination was well tolerated, and the adverse event profile was in line with previous findings.
Collapse
Affiliation(s)
- Jorge Cortes
- Department of Leukemia, Anderson Cancer Center, University of Texas, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Nikolai Podoltsev
- Department of Internal Medicine, Hematology Section, Yale University School of Medicine, New Haven, CT, USA
| | - Hagop Kantarjian
- Department of Leukemia, Anderson Cancer Center, University of Texas, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Gautam Borthakur
- Department of Leukemia, Anderson Cancer Center, University of Texas, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Amer M Zeidan
- Department of Internal Medicine, Hematology Section, Yale University School of Medicine, New Haven, CT, USA
| | - Maximilian Stahl
- Department of Internal Medicine, Hematology Section, Yale University School of Medicine, New Haven, CT, USA
| | - Tillmann Taube
- Boehringer Ingelheim International GmbH, Biberach, Germany
| | - Nora Fagan
- Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| | | | - Geoffrey L Uy
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
16
|
Alverez CN, Park JE, Toti KS, Xia Y, Krausz KW, Rai G, Bang JK, Gonzalez FJ, Jacobson KA, Lee KS. Identification of a New Heterocyclic Scaffold for Inhibitors of the Polo-Box Domain of Polo-like Kinase 1. J Med Chem 2020; 63:14087-14117. [PMID: 33175530 PMCID: PMC7769008 DOI: 10.1021/acs.jmedchem.0c01669] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
As a mitotic-specific target widely deregulated in various human cancers, polo-like kinase 1 (Plk1) has been extensively explored for anticancer activity and drug discovery. Although multiple catalytic domain inhibitors were tested in preclinical and clinical studies, their efficacies are limited by dose-limiting cytotoxicity, mainly from off-target cross reactivity. The C-terminal noncatalytic polo-box domain (PBD) of Plk1 has emerged as an attractive target for generating new protein-protein interaction inhibitors. Here, we identified a 1-thioxo-2,4-dihydro-[1,2,4]triazolo[4,3-a]quinazolin-5(1H)-one scaffold that efficiently inhibits Plk1 PBD but not its related Plk2 and Plk3 PBDs. Structure-activity relationship studies led to multiple inhibitors having ≥10-fold higher inhibitory activity than the previously characterized Plk1 PBD-specific phosphopeptide, PLHSpT (Kd ∼ 450 nM). In addition, S-methyl prodrugs effectively inhibited mitotic progression and cell proliferation and their metabolic stability was determined. These data describe a novel class of small-molecule inhibitors that offer a promising avenue for future drug discovery against Plk1-addicted cancers.
Collapse
Affiliation(s)
- Celeste N Alverez
- Chemistry Section, Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Jung-Eun Park
- Chemistry Section, Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Kiran S Toti
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Yangliu Xia
- Chemistry Section, Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Kristopher W Krausz
- Chemistry Section, Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Ganesha Rai
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Jeong K Bang
- Division of Magnetic Resonance, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Frank J Gonzalez
- Chemistry Section, Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Kyung S Lee
- Chemistry Section, Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
17
|
Kirtonia A, Pandya G, Sethi G, Pandey AK, Das BC, Garg M. A comprehensive review of genetic alterations and molecular targeted therapies for the implementation of personalized medicine in acute myeloid leukemia. J Mol Med (Berl) 2020; 98:1069-1091. [PMID: 32620999 DOI: 10.1007/s00109-020-01944-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/18/2020] [Accepted: 06/22/2020] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukemia (AML) is an extremely heterogeneous disease defined by the clonal growth of myeloblasts/promyelocytes not only in the bone marrow but also in peripheral blood and/or tissues. Gene mutations and chromosomal abnormalities are usually associated with aberrant proliferation and/or block in the normal differentiation of hematopoietic cells. So far, the combination of cytogenetic profiling and molecular and gene mutation analyses remains an essential tool for the classification, diagnosis, prognosis, and treatment for AML. This review gives an overview on how the development of novel innovative technologies has allowed us not only to detect the genetic alterations as early as possible but also to understand the molecular pathogenesis of AML to develop novel targeted therapies. We also discuss the remarkable advances made during the last decade to understand the AML genome both at primary and relapse diseases and how genetic alterations might influence the distinct biological groups as well as the clonal evolution of disease during the diagnosis and relapse. Also, the review focuses on how the persistence of epigenetic gene mutations during morphological remission is associated with relapse. It is suggested that along with the prognostic and therapeutic mutations, the novel molecular targeted therapies either approved by FDA or those under clinical trials including CART-cell therapy would be of immense importance in the effective management of AML.
Collapse
Affiliation(s)
- Anuradha Kirtonia
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India
| | - Gouri Pandya
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Amit Kumar Pandey
- Amity Institute of Biotechnology (AIB), Amity University, Gurgaon, Haryana, 122413, India
| | - Bhudev C Das
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India.
| |
Collapse
|
18
|
Identification of PLK1 as a New Therapeutic Target in Mucinous Ovarian Carcinoma. Cancers (Basel) 2020; 12:cancers12030672. [PMID: 32183025 PMCID: PMC7140026 DOI: 10.3390/cancers12030672] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/02/2020] [Accepted: 03/11/2020] [Indexed: 01/04/2023] Open
Abstract
Mucinous epithelial ovarian cancer (mEOC) is a rare subset of epithelial ovarian cancer. When diagnosed at a late stage, its prognosis is very poor, as it is quite chemo-resistant. To find new therapeutic options for mEOC, we performed high-throughput screening using a siRNA library directed against human protein kinases in a mEOC cell line, and polo-like kinase1 (PLK1) was identified as the kinase whose downregulation interfered with cell proliferation. Both PLK1 siRNA and two specific PLK1 inhibitors (onvansertib and volasertib) were able to inhibit cell growth, induce apoptosis and block cells in the G2/M phase of the cell cycle. We evaluated, in vitro, the combinations of PLK1 inhibitors and different chemotherapeutic drugs currently used in the treatment of mEOC, and we observed a synergistic effect of PLK1 inhibitors and antimitotic drugs. When translated into an in vivo xenograft model, the combination of onvansertib and paclitaxel resulted in stronger tumor regressions and in a longer mice survival than the single treatments. These effects were associated with a higher induction of mitotic block and induction of apoptosis, similarly to what was observed in vitro. These data suggest that the combination onvansertib/paclitaxel could represent a new active therapeutic option in mEOC.
Collapse
|
19
|
Hofmann MH, Mani R, Engelhardt H, Impagnatiello MA, Carotta S, Kerenyi M, Lorenzo-Herrero S, Böttcher J, Scharn D, Arnhof H, Zoephel A, Schnitzer R, Gerstberger T, Sanderson MP, Rajgolikar G, Goswami S, Vasu S, Ettmayer P, Gonzalez S, Pearson M, McConnell DB, Kraut N, Muthusamy N, Moll J. Selective and Potent CDK8/19 Inhibitors Enhance NK-Cell Activity and Promote Tumor Surveillance. Mol Cancer Ther 2020; 19:1018-1030. [PMID: 32024684 DOI: 10.1158/1535-7163.mct-19-0789] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/18/2019] [Accepted: 01/28/2020] [Indexed: 12/14/2022]
Abstract
Natural killer (NK) cells play a pivotal role in controlling cancer. Multiple extracellular receptors and internal signaling nodes tightly regulate NK activation. Cyclin-dependent kinases of the mediator complex (CDK8 and CDK19) were described as a signaling intermediates in NK cells. Here, we report for the first time the development and use of CDK8/19 inhibitors to suppress phosphorylation of STAT1S727 in NK cells and to augment the production of the cytolytic molecules perforin and granzyme B (GZMB). Functionally, this resulted in enhanced NK-cell-mediated lysis of primary leukemia cells. Treatment with the CDK8/19 inhibitor BI-1347 increased the response rate and survival of mice bearing melanoma and breast cancer xenografts. In addition, CDK8/19 inhibition augmented the antitumoral activity of anti-PD-1 antibody and SMAC mimetic therapy, both agents that promote T-cell-mediated antitumor immunity. Treatment with the SMAC mimetic compound BI-8382 resulted in an increased number of NK cells infiltrating EMT6 tumors. Combination of the CDK8/19 inhibitor BI-1347, which augments the amount of degranulation enzymes, with the SMAC mimetic BI-8382 resulted in increased survival of mice carrying the EMT6 breast cancer model. The observed survival benefit was dependent on an intermittent treatment schedule of BI-1347, suggesting the importance of circumventing a hyporesponsive state of NK cells. These results suggest that CDK8/19 inhibitors can be combined with modulators of the adaptive immune system to inhibit the growth of solid tumors, independent of their activity on cancer cells, but rather through promoting NK-cell function.
Collapse
Affiliation(s)
| | - Rajeswaran Mani
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | | | | | - Marc Kerenyi
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Seila Lorenzo-Herrero
- Department of Functional Biology, Universidad de Oviedo, Instituto de Investigación Biosanitaria del Principado de Asturias (IISPA), IUOPA, Oviedo, Spain
| | - Jark Böttcher
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Dirk Scharn
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | | | | | | | | | | - Girish Rajgolikar
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Swagata Goswami
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Sumithira Vasu
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | - Segundo Gonzalez
- Department of Functional Biology, Universidad de Oviedo, Instituto de Investigación Biosanitaria del Principado de Asturias (IISPA), IUOPA, Oviedo, Spain
| | - Mark Pearson
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | | - Norbert Kraut
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Jürgen Moll
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| |
Collapse
|
20
|
Dill V, Kauschinger J, Hauch RT, Buschhorn L, Odinius TO, Müller-Thomas C, Mishra R, Kyncl MC, Schmidt B, Prodinger PM, Hempel D, Bellos F, Höllein A, Kern W, Haferlach T, Slotta-Huspenina J, Bassermann F, Peschel C, Götze KS, Waizenegger IC, Höckendorf U, Jost PJ, Jilg S. Inhibition of PLK1 by capped-dose volasertib exerts substantial efficacy in MDS and sAML while sparing healthy haematopoiesis. Eur J Haematol 2020; 104:125-137. [PMID: 31758597 DOI: 10.1111/ejh.13354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/06/2019] [Accepted: 11/08/2019] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Targeting the cell cycle machinery represents a rational therapeutic approach in myelodysplastic syndromes (MDS) and secondary acute myeloid leukemia (sAML). Despite substantial response rates, clinical use of the PLK inhibitor volasertib has been hampered by elevated side effects such as neutropenia and infections. OBJECTIVES The primary objective was to analyse whether a reduced dose of volasertib was able to limit toxic effects on the healthy haematopoiesis while retaining its therapeutic effect. METHODS Bone marrow mononuclear cells (BMMNCs) of patients with MDS/sAML (n = 73) and healthy controls (n = 28) were treated with volasertib (1 μM to 1 nM) or vehicle control. Short-term viability analysis was performed by flow cytometry after 72 hours. For long-term viability analysis, colony-forming capacity was assessed after 14 days. Protein expression of RIPK3 and MCL-1 was quantified via flow cytometry. RESULTS Reduced dose levels of volasertib retained high cell death-inducing efficacy in primary human stem and progenitor cells of MDS/sAML patients without affecting healthy haematopoiesis in vitro. Interestingly, volasertib reduced colony-forming capacity and cell survival independent of clinical stage or mutational status. CONCLUSIONS Volasertib offers a promising therapeutic approach in patients with adverse prognostic profile. RIPK3 and MCL-1 might be potential biomarkers for sensitivity to volasertib treatment.
Collapse
Affiliation(s)
- Veronika Dill
- Medical Department III for Haematology and Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Johanna Kauschinger
- Medical Department III for Haematology and Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Richard T Hauch
- Medical Department III for Haematology and Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Lars Buschhorn
- Medical Department III for Haematology and Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Timo O Odinius
- Medical Department III for Haematology and Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Catharina Müller-Thomas
- Medical Department III for Haematology and Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Ritu Mishra
- Institute of Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Michele C Kyncl
- Medical Department III for Haematology and Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | | | - Peter M Prodinger
- Department of Orthopedic Surgery, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Dirk Hempel
- Onkologiezentrum Donauwörth, Donauworth, Germany
| | | | | | | | | | - Julia Slotta-Huspenina
- Institute of Pathology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Florian Bassermann
- Medical Department III for Haematology and Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- German Cancer Consortium (DKTK), DKFZ, Heidelberg, Germany
| | - Christian Peschel
- Medical Department III for Haematology and Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Katharina S Götze
- Medical Department III for Haematology and Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | | | - Ulrike Höckendorf
- Medical Department III for Haematology and Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Philipp J Jost
- Medical Department III for Haematology and Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- German Cancer Consortium (DKTK), DKFZ, Heidelberg, Germany
| | - Stefanie Jilg
- Medical Department III for Haematology and Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
21
|
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
|
22
|
Gatz SA, Aladowicz E, Casanova M, Chisholm JC, Kearns PR, Fulda S, Geoerger B, Schäfer BW, Shipley JM. A Perspective on Polo-Like Kinase-1 Inhibition for the Treatment of Rhabdomyosarcomas. Front Oncol 2019; 9:1271. [PMID: 31824851 PMCID: PMC6882953 DOI: 10.3389/fonc.2019.01271] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022] Open
Abstract
Rhabdomyosarcomas are the most common pediatric soft tissue sarcoma and are a major cause of death from cancer in young patients requiring new treatment options to improve outcomes. High-risk patients include those with metastatic or relapsed disease and tumors with PAX3-FOXO1 fusion genes that encode a potent transcription factor that drives tumourigenesis through transcriptional reprogramming. Polo-Like Kinase-1 (PLK1) is a serine/threonine kinase that phosphorylates a wide range of target substrates and alters their activity. PLK1 functions as a pleiotropic master regulator of mitosis and regulates DNA replication after stress. Taken together with high levels of expression that correlate with poor outcomes in many cancers, including rhabdomyosarcomas, it is an attractive therapeutic target. This is supported in rhabdomyosarcoma models by characterization of molecular and phenotypic effects of reducing and inhibiting PLK1, including changes to the PAX3-FOXO1 fusion protein. However, as tumor re-growth has been observed, combination strategies are required. Here we review preclinical evidence and consider biological rationale for PLK1 inhibition in combination with drugs that promote apoptosis, interfere with activity of PAX3-FOXO1 and are synergistic with microtubule-destabilizing drugs such as vincristine. The preclinical effects of low doses of the PLK1 inhibitor volasertib in combination with vincristine, which is widely used in rhabdomyosarcoma treatment, show particular promise in light of recent clinical data in the pediatric setting that support achievable volasertib doses predicted to be effective. Further development of novel therapeutic strategies including PLK1 inhibition may ultimately benefit young patients with rhabdomyosarcoma and other cancers.
Collapse
Affiliation(s)
- Susanne A. Gatz
- Cancer Research UK Clinical Trials Unit (CRCTU), Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Ewa Aladowicz
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | | | - Julia C. Chisholm
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Pamela R. Kearns
- Cancer Research UK Clinical Trials Unit (CRCTU), Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt, Germany
| | - Birgit Geoerger
- Gustave Roussy Cancer Campus, Department of Paediatric and Adolescent Oncology, Université Paris-Saclay, Villejuif, France
| | - Beat W. Schäfer
- Department of Oncology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Janet M. Shipley
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
23
|
Volasertib preclinical activity in high-risk hepatoblastoma. Oncotarget 2019; 10:6403-6417. [PMID: 31741706 PMCID: PMC6849653 DOI: 10.18632/oncotarget.27237] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/12/2019] [Indexed: 12/17/2022] Open
Abstract
Relapsed and metastatic hepatoblastoma represents an unmet clinical need with limited chemotherapy treatment options. In a chemical screen, we identified volasertib as an agent with in vitro activity, inhibiting hepatoblastoma cell growth while sparing normal hepatocytes. Volasertib targets PLK1 and prevents the progression of mitosis, resulting in eventual cell death. PLK1 is overexpressed in hepatoblastoma biopsies relative to normal liver tissue. As a potential therapeutic strategy, we tested the combination of volasertib and the relapse-related hepatoblastoma chemotherapeutic irinotecan. We found both in vitro and in vivo efficacy of this combination, which may merit further preclinical investigation and exploration for a clinical trial concept.
Collapse
|
24
|
Lin SF, Lin JD, Yeh CN, Huang YT, Chou TC, Wong RJ. Targeting PLKs as a therapeutic approach to well-differentiated thyroid cancer. Endocr Relat Cancer 2019; 26:727-738. [PMID: 31189135 PMCID: PMC7475022 DOI: 10.1530/erc-18-0555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 06/11/2019] [Indexed: 12/19/2022]
Abstract
Polo-like kinases (PLKs) are pivotal regulators of cell proliferation and cell survival; therefore, PLKs may be potential targets in the treatment of malignancy. The therapeutic effects of volasertib, a PLKs inhibitor for papillary and follicular thyroid cancer (known as well-differentiated thyroid cancer (WDTC)), were evaluated in this study. Volasertib inhibited cell proliferation in two papillary and two follicular thyroid cancer cell lines in a dose-dependent manner. Volasertib treatment reduced cells in the S phase and increased cells in the G2/M phase. Volasertib activated caspase-3 activity and induced apoptosis. Drug combinations of volasertib and sorafenib showed mostly synergism in four well-differentiated thyroid carcinoma cell lines in vitro. Volasertib treatment in vivo retarded the growth of a papillary thyroid tumor model. Furthermore, the combination of volasertib with sorafenib was more effective than a single treatment of either in a follicular thyroid cancer xenograft model. Promising safety profiles appeared in animals treated with either volasertib alone or volasertib and sorafenib combination therapy. These findings support volasertib as a potential drug for the treatment of patients with WDTC.
Collapse
Affiliation(s)
- Shu-Fu Lin
- Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Chang Gung University, Taoyuan, Taiwan
| | - Jen-Der Lin
- Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Chang Gung University, Taoyuan, Taiwan
| | - Chun-Nan Yeh
- Chang Gung University, Taoyuan, Taiwan
- Department of Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yu-Tung Huang
- Center for Big Data Analytics and Statistics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ting-Chao Chou
- Laboratory of Preclinical Pharmacology Core, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Current address: PD Science, Inc., 599 Mill Run, Paramus, NJ, USA
| | - Richard J. Wong
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
25
|
Van den Bossche J, Deben C, De Pauw I, Lambrechts H, Hermans C, Deschoolmeester V, Jacobs J, Specenier P, Pauwels P, Vermorken JB, Peeters M, Lardon F, Wouters A. In vitro study of the Polo-like kinase 1 inhibitor volasertib in non-small-cell lung cancer reveals a role for the tumor suppressor p53. Mol Oncol 2019; 13:1196-1213. [PMID: 30859681 PMCID: PMC6487694 DOI: 10.1002/1878-0261.12477] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 12/29/2022] Open
Abstract
Polo-like kinase 1 (Plk1), a master regulator of mitosis and the DNA damage response, is considered to be an intriguing target in the research field of mitotic intervention. The observation that Plk1 is overexpressed in multiple human malignancies, including non-small-cell lung cancer (NSCLC), gave rise to the development of several small-molecule inhibitors. Volasertib, presently the most extensively studied Plk1 inhibitor, has been validated to efficiently reduce tumor growth in preclinical settings. Unfortunately, only modest antitumor activity against solid tumors was reported in clinical trials. This discrepancy prompted research into the identification of predictive biomarkers. In this study, we investigated the therapeutic effect of volasertib monotherapy (i.e., cytotoxicity, cell cycle distribution, apoptotic cell death, cellular senescence, and migration) in a panel of NSCLC cell lines differing in p53 status under both normal and reduced oxygen tension (<0.1% O2 ). A strong growth inhibitory effect was observed in p53 wild-type cells (A549 and A549-NTC), with IC50 values significantly lower than those in p53 knockdown/mutant cells (A549-920 and NCI-H1975) (P < 0.001). While mitotic arrest was significantly greater in cells with nonfunctional p53 (P < 0.005), apoptotic cell death (P < 0.026) and cellular senescence (P < 0.021) were predominantly induced in p53 wild-type cells. Overall, the therapeutic effect of volasertib was reduced under hypoxia (P < 0.050). Remarkably, volasertib inhibited cell migration in all cell lines tested (P < 0.040), with the exception of for the NCI-H1975 p53 mutant cell line. In conclusion, our results show an important difference in the therapeutic effect of Plk1 inhibition in NSCLC cells with versus without functional p53. Overall, the p53 wild-type cell lines were more sensitive to volasertib treatment, suggesting that p53 might be a predictive biomarker for Plk1 inhibition in NSCLC. Moreover, our results pave the way for new combination strategies with Plk1 inhibitors to enhance antitumor activity.
Collapse
Affiliation(s)
| | - Christophe Deben
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
| | - Ines De Pauw
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
| | - Hilde Lambrechts
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
| | - Christophe Hermans
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
- Department of PathologyAntwerp University HospitalEdegemBelgium
| | - Vanessa Deschoolmeester
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
- Department of PathologyAntwerp University HospitalEdegemBelgium
| | - Julie Jacobs
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
- Department of PathologyAntwerp University HospitalEdegemBelgium
| | - Pol Specenier
- Department of OncologyAntwerp University HospitalEdegemBelgium
| | - Patrick Pauwels
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
- Department of PathologyAntwerp University HospitalEdegemBelgium
| | - Jan Baptist Vermorken
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
- Department of OncologyAntwerp University HospitalEdegemBelgium
| | - Marc Peeters
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
- Department of OncologyAntwerp University HospitalEdegemBelgium
| | - Filip Lardon
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
| | - An Wouters
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
| |
Collapse
|
26
|
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: 4.0] [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
|
27
|
Del Rosario BC, Kriz AJ, Del Rosario AM, Anselmo A, Fry CJ, White FM, Sadreyev RI, Lee JT. Exploration of CTCF post-translation modifications uncovers Serine-224 phosphorylation by PLK1 at pericentric regions during the G2/M transition. eLife 2019; 8:e42341. [PMID: 30676316 PMCID: PMC6361588 DOI: 10.7554/elife.42341] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/23/2019] [Indexed: 01/05/2023] Open
Abstract
The zinc finger CCCTC-binding protein (CTCF) carries out many functions in the cell. Although previous studies sought to explain CTCF multivalency based on sequence composition of binding sites, few examined how CTCF post-translational modification (PTM) could contribute to function. Here, we performed CTCF mass spectrometry, identified a novel phosphorylation site at Serine 224 (Ser224-P), and demonstrate that phosphorylation is carried out by Polo-like kinase 1 (PLK1). CTCF Ser224-P is chromatin-associated, mapping to at least a subset of known CTCF sites. CTCF Ser224-P accumulates during the G2/M transition of the cell cycle and is enriched at pericentric regions. The phospho-obviation mutant, S224A, appeared normal. However, the phospho-mimic mutant, S224E, is detrimental to mouse embryonic stem cell colonies. While ploidy and chromatin architecture appear unaffected, S224E mutants differentially express hundreds of genes, including p53 and p21. We have thus identified a new CTCF PTM and provided evidence of biological function.
Collapse
Affiliation(s)
- Brian C Del Rosario
- Department of Molecular BiologyHoward Hughes Medical Institute, Massachusetts General HospitalBostonUnited States
- Department of GeneticsHarvard Medical SchoolBostonUnited States
| | - Andrea J Kriz
- Department of Molecular BiologyHoward Hughes Medical Institute, Massachusetts General HospitalBostonUnited States
- Department of GeneticsHarvard Medical SchoolBostonUnited States
| | - Amanda M Del Rosario
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeUnited States
| | - Anthony Anselmo
- Department of Molecular BiologyMassachusetts General HospitalBostonUnited States
| | | | - Forest M White
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeUnited States
| | - Ruslan I Sadreyev
- Department of Molecular BiologyMassachusetts General HospitalBostonUnited States
| | - Jeannie T Lee
- Department of Molecular BiologyHoward Hughes Medical Institute, Massachusetts General HospitalBostonUnited States
- Department of GeneticsHarvard Medical SchoolBostonUnited States
| |
Collapse
|
28
|
Han Y, Lindner S, Bei Y, Garcia HD, Timme N, Althoff K, Odersky A, Schramm A, Lissat A, Künkele A, Deubzer HE, Eggert A, Schulte JH, Henssen AG. Synergistic activity of BET inhibitor MK-8628 and PLK inhibitor Volasertib in preclinical models of medulloblastoma. Cancer Lett 2019; 445:24-33. [PMID: 30611741 DOI: 10.1016/j.canlet.2018.12.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/05/2018] [Accepted: 12/11/2018] [Indexed: 11/20/2022]
Abstract
Medulloblastoma is the most prevalent central nervous system tumor in children. Targeted treatment approaches for patients with high-risk medulloblastoma are needed as current treatment regimens are not curative in many cases and cause significant therapy-related morbidity. Medulloblastoma harboring MYC amplification have the most aggressive clinical course and worst outcome. Targeting the BET protein BRD4 has significant anti-tumor effects in preclinical models of MYC-amplified medulloblastoma, however, in most cases these are not curative. We here assessed the therapeutic efficacy of the orally bioavailable BRD4 inhibitor, MK-8628, in preclinical models of medulloblastoma. MK-8628 showed therapeutic efficacy against in vitro and in vivo models of MYC-amplified medulloblastoma by inducing apoptotic cell death and cell cycle arrest. Gene expression analysis of cells treated with MK-8628 showed that anti-tumor effects were accompanied by significant repression of MYC transcription as well as disruption of MYC-regulated transcriptional programs. Additionally, we found that targeting of MYC protein stability through pharmacological PLK1 inhibition showed synergistic anti-medulloblastoma effects when combined with MK-8628 treatment. Thus, MK-8628 is effective against preclinical high-risk medulloblastoma models and its effects can be enhanced through simultaneous targeting of PLK1.
Collapse
Affiliation(s)
- Youjia Han
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Germany
| | - Sven Lindner
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Essen, Germany
| | - Yi Bei
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Germany
| | | | - Natalie Timme
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Germany
| | - Kristina Althoff
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Essen, Germany
| | - Andrea Odersky
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Essen, Germany
| | - Alexander Schramm
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Essen, Germany
| | - Andrej Lissat
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Germany
| | - Annette Künkele
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Germany; German Consortium for Translational Cancer Research (DKTK), Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany
| | - Hedwig E Deubzer
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Germany; German Consortium for Translational Cancer Research (DKTK), Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany; Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Germany; German Consortium for Translational Cancer Research (DKTK), Berlin, Germany
| | - Johannes H Schulte
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Germany; German Consortium for Translational Cancer Research (DKTK), Berlin, Germany
| | - Anton G Henssen
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Germany; German Consortium for Translational Cancer Research (DKTK), Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany.
| |
Collapse
|
29
|
Ottmann OG, Müller-Tidow C, Krämer A, Schlenk RF, Lübbert M, Bug G, Krug U, Bochtler T, Voss F, Taube T, Liu D, Garin-Chesa P, Döhner H. Phase I dose-escalation trial investigating volasertib as monotherapy or in combination with cytarabine in patients with relapsed/refractory acute myeloid leukaemia. Br J Haematol 2018; 184:1018-1021. [PMID: 29882583 DOI: 10.1111/bjh.15204] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Oliver G Ottmann
- Department of Medicine, Hematology/Oncology, Goethe University, Frankfurt, Germany
| | - Carsten Müller-Tidow
- Department of Medicine, Hematology and Oncology, University Hospital Münster, Münster, Germany
| | - Alwin Krämer
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Richard F Schlenk
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Michael Lübbert
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center, Freiburg, Germany
| | - Gesine Bug
- Department of Medicine, Hematology/Oncology, Goethe University, Frankfurt, Germany
| | - Utz Krug
- Department of Medicine, Hematology and Oncology, University Hospital Münster, Münster, Germany
| | - Tilmann Bochtler
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Florian Voss
- Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim, Germany
| | - Tillmann Taube
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Dan Liu
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Hartmut Döhner
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| |
Collapse
|
30
|
Gerlach D, Tontsch-Grunt U, Baum A, Popow J, Scharn D, Hofmann MH, Engelhardt H, Kaya O, Beck J, Schweifer N, Gerstberger T, Zuber J, Savarese F, Kraut N. The novel BET bromodomain inhibitor BI 894999 represses super-enhancer-associated transcription and synergizes with CDK9 inhibition in AML. Oncogene 2018; 37:2687-2701. [PMID: 29491412 PMCID: PMC5955861 DOI: 10.1038/s41388-018-0150-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/22/2017] [Accepted: 12/30/2017] [Indexed: 01/12/2023]
Abstract
Bromodomain and extra-terminal (BET) protein inhibitors have been reported as treatment options for acute myeloid leukemia (AML) in preclinical models and are currently being evaluated in clinical trials. This work presents a novel potent and selective BET inhibitor (BI 894999), which has recently entered clinical trials (NCT02516553). In preclinical studies, this compound is highly active in AML cell lines, primary patient samples, and xenografts. HEXIM1 is described as an excellent pharmacodynamic biomarker for target engagement in tumors as well as in blood. Mechanistic studies show that BI 894999 targets super-enhancer-regulated oncogenes and other lineage-specific factors, which are involved in the maintenance of the disease state. BI 894999 is active as monotherapy in AML xenografts, and in addition leads to strongly enhanced antitumor effects in combination with CDK9 inhibitors. This treatment combination results in a marked decrease of global p-Ser2 RNA polymerase II levels and leads to rapid induction of apoptosis in vitro and in vivo. Together, these data provide a strong rationale for the clinical evaluation of BI 894999 in AML.
Collapse
Affiliation(s)
- Daniel Gerlach
- Boehringer Ingelheim RCV GmbH & Co KG, 1120, Vienna, Austria
| | | | - Anke Baum
- Boehringer Ingelheim RCV GmbH & Co KG, 1120, Vienna, Austria
| | - Johannes Popow
- Boehringer Ingelheim RCV GmbH & Co KG, 1120, Vienna, Austria
| | - Dirk Scharn
- Boehringer Ingelheim RCV GmbH & Co KG, 1120, Vienna, Austria
| | - Marco H Hofmann
- Boehringer Ingelheim RCV GmbH & Co KG, 1120, Vienna, Austria
| | | | - Onur Kaya
- Boehringer Ingelheim RCV GmbH & Co KG, 1120, Vienna, Austria
| | - Janina Beck
- Boehringer Ingelheim RCV GmbH & Co KG, 1120, Vienna, Austria
| | | | | | - Johannes Zuber
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030, Vienna, Austria.,Medical University of Vienna, Vienna BioCenter (VBC), 1030, Vienna, Austria
| | - Fabio Savarese
- Boehringer Ingelheim RCV GmbH & Co KG, 1120, Vienna, Austria.
| | - Norbert Kraut
- Boehringer Ingelheim RCV GmbH & Co KG, 1120, Vienna, Austria.
| |
Collapse
|
31
|
Tontsch-Grunt U, Rudolph D, Waizenegger I, Baum A, Gerlach D, Engelhardt H, Wurm M, Savarese F, Schweifer N, Kraut N. Synergistic activity of BET inhibitor BI 894999 with PLK inhibitor volasertib in AML in vitro and in vivo. Cancer Lett 2018; 421:112-120. [PMID: 29454094 DOI: 10.1016/j.canlet.2018.02.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 01/29/2018] [Accepted: 02/11/2018] [Indexed: 12/11/2022]
Abstract
Interactions between a new potent Bromodomain and extraterminal domain (BET) inhibitor BI 894999 and the polo-like kinase (PLK) inhibitor volasertib were studied in acute myeloid leukemia cell lines in vitro and in vivo. We provide data for the distinct mechanisms of action of these two compounds with a potential utility in AML based on gene expression, cell cycle profile and modulation of PD biomarkers such as MYC and HEXIM1. In contrast to BI 894999, volasertib treatment neither affects MYC nor HEXIM1 expression, but augments and prolongs the decrease of MYC expression caused by BI 894999 treatment. In vitro combination of both compounds leads to a decrease in S-Phase and to increased apoptosis. In vitro scheduling experiments guided in vivo experiments in disseminated AML mouse models. Co-administration of BI 894999 and volasertib dramatically reduces tumor burden accompanied by long-term survival of tumor-bearing mice and eradication of AML cells in mouse bone marrow. Together, these preclinical findings provide evidence for the strong synergistic effect of BI 894999 and volasertib, warranting future clinical studies in patients with AML to investigate this paradigm.
Collapse
Affiliation(s)
| | | | | | - Anke Baum
- Boehringer Ingelheim RCV GmbH & Co KG, A-1120 Vienna, Austria
| | - Daniel Gerlach
- Boehringer Ingelheim RCV GmbH & Co KG, A-1120 Vienna, Austria
| | | | - Melanie Wurm
- Boehringer Ingelheim RCV GmbH & Co KG, A-1120 Vienna, Austria
| | - Fabio Savarese
- Boehringer Ingelheim RCV GmbH & Co KG, A-1120 Vienna, Austria
| | | | - Norbert Kraut
- Boehringer Ingelheim RCV GmbH & Co KG, A-1120 Vienna, Austria
| |
Collapse
|
32
|
Gopalakrishnan B, Cheney C, Mani R, Mo X, Bucci D, Walker A, Klisovic R, Bhatnagar B, Walsh K, Rueter B, Waizenegger IC, Heider KH, Blum W, Vasu S, Muthusamy N. Polo-like kinase inhibitor volasertib marginally enhances the efficacy of the novel Fc-engineered anti-CD33 antibody BI 836858 in acute myeloid leukemia. Oncotarget 2018. [PMID: 29515764 PMCID: PMC5839395 DOI: 10.18632/oncotarget.23880] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Acute myeloid leukemia (AML) is the second most common type of leukemia in adults. Incidence of AML increases with age with a peak incidence at 67 years. Patients older than 60 years have an unfavorable prognosis due to resistance to conventional chemotherapy. Volasertib (BI 6727) is a cell-cycle regulator targeting polo-like kinase which has been evaluated in clinical trials in AML. We evaluated effects of volasertib in primary patient samples and NK cells. At equivalent doses, volasertib is cytotoxic to AML blasts but largely spares healthy NK cells. We then evaluated the effect of volasertib treatment in combination with BI 836858 on primary AML blast samples using antibody-dependent cellular cytotoxicity (ADCC) assays. Volasertib treatment of NK cells did not impair NK function as evidenced by comparable levels of BI 836858 mediated ADCC in both volasertib-treated and control-treated NK cells. In summary, volasertib is cytotoxic to AML blasts while sparing NK cell viability and function. Higher BI 836858 mediated ADCC was observed in patient samples pretreated with volasertib. These findings provide a strong rationale to test combination of BI 836858 and volasertib in AML.
Collapse
Affiliation(s)
| | - Carolyn Cheney
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Rajeswaran Mani
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Xiaokui Mo
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Donna Bucci
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Alison Walker
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Rebecca Klisovic
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Bhavana Bhatnagar
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Katherine Walsh
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Bjoern Rueter
- Boehringer Ingelheim Pharma GmbH, Biberach/Riss, Germany
| | | | | | - William Blum
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Sumithira Vasu
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Natarajan Muthusamy
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
33
|
Alimova I, Pierce AM, Harris P, Donson A, Birks DK, Prince E, Balakrishnan I, Foreman NK, Kool M, Hoffman L, Venkataraman S, Vibhakar R. Targeting Polo-like kinase 1 in SMARCB1 deleted atypical teratoid rhabdoid tumor. Oncotarget 2017; 8:97290-97303. [PMID: 29228610 PMCID: PMC5722562 DOI: 10.18632/oncotarget.21932] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 08/15/2017] [Indexed: 12/15/2022] Open
Abstract
Atypical teratoid rhabdoid tumor (ATRT) is an aggressive and malignant pediatric brain tumor. Polo-like kinase 1 (PLK1) is highly expressed in many cancers and essential for mitosis. Overexpression of PLK1 promotes chromosome instability and aneuploidy by overriding the G2-M DNA damage and spindle checkpoints. Recent studies suggest that targeting PLK1 by small molecule inhibitors is a promising approach to tumor therapy. We investigated the effect of PLK1 inhibition in ATRT. Gene expression analysis showed that PLK1 was overexpressed in ATRT patient samples and tumor cell lines. Genetic inhibition of PLK1 with shRNA potently suppressed ATRT cell growth in vitro. Treatment with the PLK1 inhibitor BI 6727 (Volasertib) significantly decreased cell growth, inhibited clonogenic potential, and induced apoptosis. BI6727 treatment led to G2-M phase arrest, consistent with PLK1's role as a critical regulator of mitosis. Moreover, inhibition of PLK1 by BI6727 suppressed the tumor-sphere formation of ATRT cells. Treatment also significantly decreased levels of the DNA damage proteins Ku80 and RAD51 and increased γ-H2AX expression, indicating that BI 6727 can induce DNA damage. Importantly, BI6727 significantly enhanced radiation sensitivity of ATRT cells. In vivo, BI6727 slowed growth of ATRT tumors and prolonged survival in a xenograft model. PLK1 inhibition is a compelling new therapeutic approach for treating ATRT, and the use of BI6727 should be evaluated in clinical studies.
Collapse
Affiliation(s)
- Irina Alimova
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Angela M Pierce
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Peter Harris
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Andrew Donson
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Diane K Birks
- Department of Neurosurgery, University of Colorado Denver, Aurora, CO, United States
| | - Eric Prince
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Ilango Balakrishnan
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Nicholas K Foreman
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, CO, United States.,Department of Neurosurgery, University of Colorado Denver, Aurora, CO, United States
| | - Marcel Kool
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lindsey Hoffman
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Sujatha Venkataraman
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, CO, United States
| | - Rajeev Vibhakar
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, CO, United States.,Department of Neurosurgery, University of Colorado Denver, Aurora, CO, United States
| |
Collapse
|
34
|
Adachi Y, Ishikawa Y, Kiyoi H. Identification of volasertib-resistant mechanism and evaluation of combination effects with volasertib and other agents on acute myeloid leukemia. Oncotarget 2017; 8:78452-78465. [PMID: 29108241 PMCID: PMC5667974 DOI: 10.18632/oncotarget.19632] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/03/2017] [Indexed: 11/25/2022] Open
Abstract
Volasertib, a selective PLK1 inhibitor, was effective for acute myeloid leukemia (AML) patients in clinical trials. However, its efficacy was limited in mono-therapy, and a higher incidence of fatal events was revealed in the combination with low-dose cytarabine. Thus, optimization of combination therapy with volasertib and other agents is necessary for its clinical development, and the predictive factors for response or resistance to volasertib remain largely unknown. In this study, we investigated the resistance mechanism in volasertib-resistant cell lines and the combination effects with other agents, such as azacitidine (AZA), on AML cells. We identified that mutations in the ATP-binding domain of PLK1 and expression of MDR1 conferred resistance to volasertib. In the combination therapy, the effects of AZA differed among cells, but were prominent in the cells with higher GI50 values of volasertib in mono-therapy. Furthermore, we identified that the cells in G2/M phase were more sensitive to volasertib, and the PI3K/AKT pathway was up-regulated upon administration of volasertib. Combination therapies with the agents that caused cell cycle accumulation in G2/M phase or with PI3K inhibitor were highly potent against AML cells. Our findings provide strategies for further clinical development of volasertib and PLK inhibitors for AML.
Collapse
Affiliation(s)
- Yoshiya Adachi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yuichi Ishikawa
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|
35
|
P. Solans B, Fleury A, Freiwald M, Fritsch H, Haug K, Trocóniz IF. Population Pharmacokinetics of Volasertib Administered in Patients with Acute Myeloid Leukaemia as a Single Agent or in Combination with Cytarabine. Clin Pharmacokinet 2017. [DOI: 10.1007/s40262-017-0566-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
36
|
Abstract
Cancer is characterized by uncontrolled tumour cell proliferation resulting from aberrant activity of various cell cycle proteins. Therefore, cell cycle regulators are considered attractive targets in cancer therapy. Intriguingly, animal models demonstrate that some of these proteins are not essential for proliferation of non-transformed cells and development of most tissues. By contrast, many cancers are uniquely dependent on these proteins and hence are selectively sensitive to their inhibition. After decades of research on the physiological functions of cell cycle proteins and their relevance for cancer, this knowledge recently translated into the first approved cancer therapeutic targeting of a direct regulator of the cell cycle. In this Review, we focus on proteins that directly regulate cell cycle progression (such as cyclin-dependent kinases (CDKs)), as well as checkpoint kinases, Aurora kinases and Polo-like kinases (PLKs). We discuss the role of cell cycle proteins in cancer, the rationale for targeting them in cancer treatment and results of clinical trials, as well as the future therapeutic potential of various cell cycle inhibitors.
Collapse
Affiliation(s)
- Tobias Otto
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02215, USA
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02215, USA
| |
Collapse
|
37
|
Selective Inhibitors of Histone Deacetylases 1 and 2 Synergize with Azacitidine in Acute Myeloid Leukemia. PLoS One 2017; 12:e0169128. [PMID: 28060870 PMCID: PMC5218480 DOI: 10.1371/journal.pone.0169128] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 12/12/2016] [Indexed: 12/21/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous group of hematopoietic stem cell disorders characterized by defects in myeloid differentiation and increased proliferation of neoplastic hematopoietic precursor cells. Outcomes for patients with AML remain poor, highlighting the need for novel treatment options. Aberrant epigenetic regulation plays an important role in the pathogenesis of AML, and inhibitors of DNA methyltransferase or histone deacetylase (HDAC) enzymes have exhibited activity in preclinical AML models. Combination studies with HDAC inhibitors plus DNA methyltransferase inhibitors have potential beneficial clinical activity in AML, however the toxicity profiles of non-selective HDAC inhibitors in the combination setting limit their clinical utility. In this work, we describe the preclinical development of selective inhibitors of HDAC1 and HDAC2, which are hypothesized to have improved safety profiles, for combination therapy in AML. We demonstrate that selective inhibition of HDAC1 and HDAC2 is sufficient to achieve efficacy both as a single agent and in combination with azacitidine in preclinical models of AML, including established AML cell lines, primary leukemia cells from AML patient bone marrow samples and in vivo xenograft models of human AML. Gene expression profiling of AML cells treated with either an HDAC1/2 inhibitor, azacitidine, or the combination of both have identified a list of genes involved in transcription and cell cycle regulation as potential mediators of the combinatorial effects of HDAC1/2 inhibition with azacitidine. Together, these findings support the clinical evaluation of selective HDAC1/2 inhibitors in combination with azacitidine in AML patients.
Collapse
|
38
|
Liu Z, Sun Q, Wang X. PLK1, A Potential Target for Cancer Therapy. Transl Oncol 2016; 10:22-32. [PMID: 27888710 PMCID: PMC5124362 DOI: 10.1016/j.tranon.2016.10.003] [Citation(s) in RCA: 291] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 10/06/2016] [Accepted: 10/11/2016] [Indexed: 12/14/2022] Open
Abstract
Polo-like kinase 1 (PLK1) plays an important role in the initiation, maintenance, and completion of mitosis. Dysfunction of PLK1 may promote cancerous transformation and drive its progression. PLK1 overexpression has been found in a variety of human cancers and was associated with poor prognoses in cancers. Many studies have showed that inhibition of PLK1 could lead to death of cancer cells by interfering with multiple stages of mitosis. Thus, PLK1 is expected to be a potential target for cancer therapy. In this article, we examined PLK1’s structural characteristics, its regulatory roles in cell mitosis, PLK1 expression, and its association with survival prognoses of cancer patients in a wide variety of cancer types, PLK1 interaction networks, and PLK1 inhibitors under investigation. Finally, we discussed the key issues in the development of PLK1-targeted cancer therapy.
Collapse
Affiliation(s)
- Zhixian Liu
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qingrong Sun
- School of Science, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaosheng Wang
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
39
|
Van den Bossche J, Lardon F, Deschoolmeester V, De Pauw I, Vermorken JB, Specenier P, Pauwels P, Peeters M, Wouters A. Spotlight on Volasertib: Preclinical and Clinical Evaluation of a Promising Plk1 Inhibitor. Med Res Rev 2016; 36:749-86. [PMID: 27140825 DOI: 10.1002/med.21392] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 02/05/2016] [Accepted: 03/10/2016] [Indexed: 12/20/2022]
Abstract
Considering the important side effects of conventional microtubule targeting agents, more and more research focuses on regulatory proteins for the development of mitosis-specific agents. Polo-like kinase 1 (Plk1), a master regulator of several cell cycle events, has arisen as an intriguing target in this research field. The observed overexpression of Plk1 in a broad range of human malignancies has given rise to the development of several potent and specific small molecule inhibitors targeting the kinase. In this review, we focus on volasertib (BI6727), the lead agent in category of Plk1 inhibitors at the moment. Numerous preclinical experiments have demonstrated that BI6727 is highly active across a variety of carcinoma cell lines, and the inhibitor has been reported to induce tumor regression in several xenograft models. Moreover, volasertib has shown clinical efficacy in multiple tumor types. As a result, Food and Drug Administration (FDA) has recently awarded volasertib the Breakthrough Therapy status after significant benefit was observed in acute myeloid leukemia (AML) patients treated with the Plk1 inhibitor. Here, we discuss both preclinical and clinical data available for volasertib administered as monotherapy or in combination with other anticancer therapies in a broad range of tumor types.
Collapse
Affiliation(s)
- J Van den Bossche
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Antwerp, Belgium
| | - F Lardon
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Antwerp, Belgium
| | - V Deschoolmeester
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Antwerp, Belgium
- Department of Pathology, Antwerp University Hospital, Edegem, Belgium
| | - I De Pauw
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Antwerp, Belgium
| | - J B Vermorken
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Antwerp, Belgium
- Department of Oncology, Antwerp University Hospital, Edegem, Belgium
| | - P Specenier
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Antwerp, Belgium
- Department of Oncology, Antwerp University Hospital, Edegem, Belgium
| | - P Pauwels
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Antwerp, Belgium
- Department of Pathology, Antwerp University Hospital, Edegem, Belgium
| | - M Peeters
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Antwerp, Belgium
- Department of Oncology, Antwerp University Hospital, Edegem, Belgium
| | - A Wouters
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
40
|
Gutteridge REA, Ndiaye MA, Liu X, Ahmad N. Plk1 Inhibitors in Cancer Therapy: From Laboratory to Clinics. Mol Cancer Ther 2016; 15:1427-35. [PMID: 27330107 PMCID: PMC4936921 DOI: 10.1158/1535-7163.mct-15-0897] [Citation(s) in RCA: 269] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 04/06/2016] [Indexed: 01/06/2023]
Abstract
Polo-like kinase 1 (Plk1) overexpression has been shown to occur in a wide range of tumors, prompting research and development of Plk1 inhibitors as a means of cancer treatment. This review discusses recent advances in the development of Plk1 inhibitors for cancer management. Plk1 inhibition has been shown to cause mitotic block and apoptosis of cells with higher mitotic index and therefore higher Plk1 expression. The potential of Plk1 inhibitors as cancer therapeutics has been widely investigated. However, a complete understanding of Plk1 biology/mechanism is yet to be fully achieved. Resistance to certain chemotherapeutic drugs has been linked to Plk1 overexpression, and Plk1-mediated mitotic events such as microtubule rearrangement have been found to reduce the efficacy of chemotherapeutic agents. The Plk1 inhibitor volasertib has shown considerable promise in clinical studies, having reached phase III trials. However, preclinical success with Plk1 inhibitors has not translated well into clinical success. In our view, combined therapies targeting other relevant pathways together with Plk1 may be vital to combat issues observed with monotherapy, especially resistance. In addition, research should also be directed toward understanding the mechanisms of Plk1 and designing additional next generations of specific, potent Plk1 inhibitors to target cancer. Mol Cancer Ther; 15(7); 1427-35. ©2016 AACR.
Collapse
Affiliation(s)
| | - Mary Ann Ndiaye
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin
| | - Xiaoqi Liu
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin. William S. Middleton Memorial VA Hospital, Madison, Wisconsin.
| |
Collapse
|
41
|
Fu G, Somasundaram RT, Jessa F, Srivastava G, MacMillan C, Witterick I, Walfish PG, Ralhan R. ER maleate is a novel anticancer agent in oral cancer: implications for cancer therapy. Oncotarget 2016; 7:17162-81. [PMID: 26934445 PMCID: PMC4941378 DOI: 10.18632/oncotarget.7751] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 01/07/2016] [Indexed: 12/29/2022] Open
Abstract
ER maleate [10-(3-Aminopropyl)-3, 4-dimethyl-9(10H)-acridinone maleate] identified in a kinome screen was investigated as a novel anticancer agent for oral squamous cell carcinoma (OSCC). Our aim was to demonstrate its anticancer effects, identify putative molecular targets and determine their clinical relevance and investigate its chemosensitization potential for platinum drugs to aid in OSCC management. Biologic effects of ER maleate were determined using oral cancer cell lines in vitro and oral tumor xenografts in vivo. mRNA profiling, real time PCR and western blot revealed ER maleate modulated the expression of polo-like kinase 1 (PLK1) and spleen tyrosine kinase (Syk). Their clinical significance was determined in oral SCC patients by immunohistochemistry and correlated with prognosis by Kaplan-Meier survival and multivariate Cox regression analyses. ER maleate induced cell apoptosis, inhibited proliferation, colony formation, migration and invasion in oral cancer cells. Imagestream analysis revealed cell cycle arrest in G2/M phase and increased polyploidy, unravelling deregulation of cell division and cell death. Mechanistically, ER maleate decreased expression of PLK1 and Syk, induced cleavage of PARP, caspase9 and caspase3, and increased chemosensitivity to carboplatin; significantly suppressed tumor growth and increased antitumor activity of carboplatin in tumor xenografts. ER maleate treated tumor xenografts showed reduced PLK1 and Syk expression. Clinical investigations revealed overexpression of PLK1 and Syk in oral SCC patients that correlated with disease prognosis. Our in vitro and in vivo findings provide a strong rationale for pre-clinical efficacy of ER maleate as a novel anticancer agent and chemosensitizer of platinum drugs for OSCC.
Collapse
Affiliation(s)
- Guodong Fu
- Department of Medicine, Alex and Simona Shnaider Research Laboratory in Molecular Oncology, Endocrine Division, Mount Sinai Hospital, Toronto, Canada
| | - Raj Thani Somasundaram
- Department of Medicine, Alex and Simona Shnaider Research Laboratory in Molecular Oncology, Endocrine Division, Mount Sinai Hospital, Toronto, Canada
| | - Fatima Jessa
- Department of Medicine, Alex and Simona Shnaider Research Laboratory in Molecular Oncology, Endocrine Division, Mount Sinai Hospital, Toronto, Canada
| | - Gunjan Srivastava
- Department of Medicine, Alex and Simona Shnaider Research Laboratory in Molecular Oncology, Endocrine Division, Mount Sinai Hospital, Toronto, Canada
| | - Christina MacMillan
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
| | - Ian Witterick
- Department of Otolaryngology — Head and Neck Surgery, Joseph and Mildred Sonshine Family Centre for Head and Neck Diseases, Mount Sinai Hospital, Toronto, Canada
- Department of Otolaryngology — Head and Neck Surgery, University of Toronto, Toronto, Canada
| | - Paul G. Walfish
- Department of Medicine, Alex and Simona Shnaider Research Laboratory in Molecular Oncology, Endocrine Division, Mount Sinai Hospital, Toronto, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
- Department of Otolaryngology — Head and Neck Surgery, Joseph and Mildred Sonshine Family Centre for Head and Neck Diseases, Mount Sinai Hospital, Toronto, Canada
- Department of Medicine, Endocrine Division, Mount Sinai Hospital and University of Toronto, Toronto, Canada
- Department of Otolaryngology — Head and Neck Surgery, University of Toronto, Toronto, Canada
| | - Ranju Ralhan
- Department of Medicine, Alex and Simona Shnaider Research Laboratory in Molecular Oncology, Endocrine Division, Mount Sinai Hospital, Toronto, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
- Department of Otolaryngology — Head and Neck Surgery, Joseph and Mildred Sonshine Family Centre for Head and Neck Diseases, Mount Sinai Hospital, Toronto, Canada
- Department of Otolaryngology — Head and Neck Surgery, University of Toronto, Toronto, Canada
| |
Collapse
|
42
|
Pujade-Lauraine E, Selle F, Weber B, Ray-Coquard IL, Vergote I, Sufliarsky J, Del Campo JM, Lortholary A, Lesoin A, Follana P, Freyer G, Pardo B, Vidal L, Tholander B, Gladieff L, Sassi M, Garin-Chesa P, Nazabadioko S, Marzin K, Pilz K, Joly F. Volasertib Versus Chemotherapy in Platinum-Resistant or -Refractory Ovarian Cancer: A Randomized Phase II Groupe des Investigateurs Nationaux pour l'Etude des Cancers de l'Ovaire Study. J Clin Oncol 2016; 34:706-13. [PMID: 26755507 DOI: 10.1200/jco.2015.62.1474] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
PURPOSE Volasertib is a potent and selective cell-cycle kinase inhibitor that induces mitotic arrest and apoptosis by targeting Polo-like kinase. This phase II trial evaluated volasertib or single-agent chemotherapy in patients with platinum-resistant or -refractory ovarian cancer who experienced failure after treatment with two or three therapy lines. PATIENTS AND METHODS Patients were randomly assigned to receive either volasertib 300 mg by intravenous infusion every 3 weeks or an investigator's choice of single-agent, nonplatinum, cytotoxic chemotherapy. The primary end point was 24-week disease control rate. Secondary end points included best overall response, progression-free survival (PFS), safety, quality of life, and exploratory biomarker analyses. RESULTS Of the 109 patients receiving treatment, 54 received volasertib and 55 received chemotherapy; demographics were well balanced. The 24-week disease control rates for volasertib and chemotherapy were 30.6% (95% CI, 18.0% to 43.2%) and 43.1% (95% CI, 29.6% to 56.7%), respectively, with partial responses in seven (13.0%) and eight (14.5%) patients, respectively. Median PFS was 13.1 weeks and 20.6 weeks for volasertib and chemotherapy (hazard ratio, 1.01; 95% CI, 0.66 to 1.53). Six patients (11%) receiving volasertib achieved PFS fore more than 1 year, whereas no patient receiving chemotherapy achieved PFS greater than 1 year. No relationship between the expression of the biomarkers tested and their response was determined. Patients treated with volasertib experienced more grade 3 and 4 drug-related hematologic adverse events (AEs) and fewer nonhematologic AEs than did patients receiving chemotherapy. Discontinuation resulting from AEs occurred in seven (13.0%) and 15 (27.3%) patients in the volasertib and chemotherapy arms, respectively. Both arms showed similar effects on quality of life. CONCLUSION Single-agent volasertib showed antitumor activity in patients with ovarian cancer. AEs in patients receiving volasertib were mainly hematologic and manageable.
Collapse
Affiliation(s)
- Eric Pujade-Lauraine
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany.
| | - Frédéric Selle
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Béatrice Weber
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Isabelle-Laure Ray-Coquard
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Ignace Vergote
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Jozef Sufliarsky
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Josep Maria Del Campo
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Alain Lortholary
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Anne Lesoin
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Philippe Follana
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Gilles Freyer
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Beatriz Pardo
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Laura Vidal
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Bengt Tholander
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Laurence Gladieff
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Mouna Sassi
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Pilar Garin-Chesa
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Serge Nazabadioko
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Kristell Marzin
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Korinna Pilz
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| | - Florence Joly
- Eric Pujade-Lauraine, Centre des Cancers de la Femme et Recherche Clinique, Paris; Frédéric Selle, Hôpitaux Universitaires de l'Est Parisien-site Tenon and Alliance Pour la Recherche En Cancérologie, Paris; Béatrice Weber, Centre Alexis Vautrin, Vandoeuvre-les-Nancy; Isabelle-Laure Ray-Coquard, Centre Léon Bérard and Université Claude Bernard-Lyon I, Lyon; Alain Lortholary, Centre Catherine de Sienne, Nantes; Anne Lesoin, Centre Oscar Lambret, Lille; Philippe Follana, Centre Antoine-Lacassagne, Nice; Gilles Freyer, Lyon University, Hospices Civils de Lyon, Pierre-Bénite Cédex; Laurence Gladieff, Institut Claudius Regaud-IUCTO, Toulouse; Mouna Sassi and Serge Nazabadioko, Boehringer Ingelheim, Reims; Florence Joly, Centre François Baclesse, Caen, France; Ignace Vergote, University Hospitals Leuven, KU Leuven, Leuven, Belgium; Jozef Sufliarsky, National Cancer Institute, Bratislava, Slovakia; Josep Maria Del Campo, Hospital University, Vall d'Hebrón; Beatriz Pardo, Institut Català d'Oncologia-Instituto de Investigación Biomédica de Bellvitge; Laura Vidal, Hospital Clínic de Barcelona, Barcelona, Spain; Bengt Tholander, Uppsala University Hospital, Uppsala, Sweden; Pilar Garin-Chesa, Boehringer Ingelheim, Vienna, Austria; Kristell Marzin, Boehringer Ingelheim, Biberach; and Korinna Pilz, Boehringer Ingelheim, Ingelheim, Germany
| |
Collapse
|
43
|
Long T, Neitz RJ, Beasley R, Kalyanaraman C, Suzuki BM, Jacobson MP, Dissous C, McKerrow JH, Drewry DH, Zuercher WJ, Singh R, Caffrey CR. Structure-Bioactivity Relationship for Benzimidazole Thiophene Inhibitors of Polo-Like Kinase 1 (PLK1), a Potential Drug Target in Schistosoma mansoni. PLoS Negl Trop Dis 2016; 10:e0004356. [PMID: 26751972 PMCID: PMC4709140 DOI: 10.1371/journal.pntd.0004356] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 12/13/2015] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Schistosoma flatworm parasites cause schistosomiasis, a chronic and debilitating disease of poverty in developing countries. Praziquantel is employed for treatment and disease control. However, its efficacy spectrum is incomplete (less active or inactive against immature stages of the parasite) and there is a concern of drug resistance. Thus, there is a need to identify new drugs and drug targets. METHODOLOGY/PRINCIPAL FINDINGS We show that RNA interference (RNAi) of the Schistosoma mansoni ortholog of human polo-like kinase (huPLK)1 elicits a deleterious phenotypic alteration in post-infective larvae (schistosomula or somules). Phenotypic screening and analysis of schistosomula and adult S. mansoni with small molecule inhibitors of huPLK1 identified a number of potent anti-schistosomals. Among these was a GlaxoSmithKline (GSK) benzimidazole thiophene inhibitor that has completed Phase I clinical trials for treatment of solid tumor malignancies. We then obtained GSKs Published Kinase Inhibitor Sets (PKIS) 1 and 2, and phenotypically screened an expanded series of 38 benzimidazole thiophene PLK1 inhibitors. Computational analysis of controls and PLK1 inhibitor-treated populations of somules demonstrated a distinctive phenotype distribution. Using principal component analysis (PCA), the phenotypes exhibited by these populations were mapped, visualized and analyzed through projection to a low-dimensional space. The phenotype distribution was found to have a distinct shape and topology, which could be elicited using cluster analysis. A structure-activity relationship (SAR) was identified for the benzimidazole thiophenes that held for both somules and adult parasites. The most potent inhibitors produced marked phenotypic alterations at 1-2 μM within 1 h. Among these were compounds previously characterized as potent inhibitors of huPLK1 in cell assays. CONCLUSIONS/SIGNIFICANCE The reverse genetic and chemical SAR data support a continued investigation of SmPLK1 as a possible drug target and/or the prosecution of the benzimidazole thiophene chemotype as a source of novel anti-schistosomals.
Collapse
Affiliation(s)
- Thavy Long
- Center for Discovery and Innovation in Parasitic Diseases, University of California, San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California, San Francisco, San Francisco, California, United States of America
| | - R. Jeffrey Neitz
- Center for Discovery and Innovation in Parasitic Diseases, University of California, San Francisco, San Francisco, California, United States of America
- Small Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, United States of America
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, United States of America
| | - Rachel Beasley
- Department of Computer Science, San Francisco State University, San Francisco, California, United States of America
| | - Chakrapani Kalyanaraman
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, United States of America
| | - Brian M. Suzuki
- Center for Discovery and Innovation in Parasitic Diseases, University of California, San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California, San Francisco, San Francisco, California, United States of America
| | - Matthew P. Jacobson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, United States of America
| | - Colette Dissous
- Center of Infection and Immunity of Lille, Université Lille Nord de France, Inserm U1019, CNRS-UMR 8204, Institut Pasteur de Lille, Lille, France
| | - James H. McKerrow
- Center for Discovery and Innovation in Parasitic Diseases, University of California, San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California, San Francisco, San Francisco, California, United States of America
| | - David H. Drewry
- Department of Chemical Biology, GlaxoSmithKline, Research Triangle Park, North Carolina, United States of America
| | - William J. Zuercher
- Department of Chemical Biology, GlaxoSmithKline, Research Triangle Park, North Carolina, United States of America
| | - Rahul Singh
- Center for Discovery and Innovation in Parasitic Diseases, University of California, San Francisco, San Francisco, California, United States of America
- Department of Computer Science, San Francisco State University, San Francisco, California, United States of America
| | - Conor R. Caffrey
- Center for Discovery and Innovation in Parasitic Diseases, University of California, San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California, San Francisco, San Francisco, California, United States of America
| |
Collapse
|
44
|
Lee KS, Burke TR, Park JE, Bang JK, Lee E. Recent Advances and New Strategies in Targeting Plk1 for Anticancer Therapy. Trends Pharmacol Sci 2015; 36:858-877. [PMID: 26478211 PMCID: PMC4684765 DOI: 10.1016/j.tips.2015.08.013] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 08/21/2015] [Accepted: 08/21/2015] [Indexed: 12/11/2022]
Abstract
Polo-like kinase 1 (Plk1) plays key roles in regulating mitotic processes that are crucial for cellular proliferation. Overexpression of Plk1 is tightly associated with the development of particular cancers in humans, and a large body of evidence suggests that Plk1 is an attractive target for anticancer therapeutic development. Drugs targeting Plk1 can potentially be directed at two distinct sites: the N-terminal catalytic kinase domain (KD), which phosphorylates substrates, and the C-terminal polo-box domain (PBD) which is essential for protein-protein interactions. In this review we summarize recent advances and new challenges in the development of Plk1 inhibitors targeting these two domains. We also discuss novel strategies for designing and developing next-generation inhibitors to effectively treat Plk1-associated human disorders.
Collapse
Affiliation(s)
- Kyung S Lee
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Terrence R Burke
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Jung-Eun Park
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeong K Bang
- Division of Magnetic Resonance, Korea Basic Science Institute, 804-1, Yangcheong Ri, Ochang, Chungbuk, Cheongwon 363-883, Republic of Korea
| | - Eunhye Lee
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
45
|
Wu CP, Hsieh CH, Hsiao SH, Luo SY, Su CY, Li YQ, Huang YH, Huang CW, Hsu SC. Human ATP-Binding Cassette Transporter ABCB1 Confers Resistance to Volasertib (BI 6727), a Selective Inhibitor of Polo-like Kinase 1. Mol Pharm 2015; 12:3885-95. [PMID: 26412161 DOI: 10.1021/acs.molpharmaceut.5b00312] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The overexpression of the serine/threonine specific polo-like kinase 1 (Plk1) is associated with poor prognosis in many types of cancer. Consequently, Plk1 has emerged as a valid therapeutic target for anticancer drug design. Volasertib is a potent inhibitor of Plk1 that inhibits the proliferation of multiple human cancer cell lines by promoting cell cycle arrest at nanomolar concentrations. However, the risk of developing drug resistance, which is often associated with the overexpression of the ATP-binding cassette (ABC) transporter ABCB1 (P-glycoprotein), can present a therapeutic challenge for volasertib and many other therapeutic drugs. Although volasertib is highly effective against the proliferation of numerous cancer cell lines, we found that the overexpression of ABCB1 in cancer cells leads to cellular resistance to volasertib and reduces the level of volasertib-stimulated G2/M cell cycle arrest and subsequent onset of apoptosis. Furthermore, we demonstrate that volasertib competitively inhibits the function of ABCB1 and stimulates the basal ATPase activity of ABCB1 in a concentration-dependent manner, which is consistent with substrate transport by ABCB1. More importantly, we discovered that the coadministration of an inhibitor or drug substrate of ABCB1 restored the anticancer activity of volasertib in ABCB1-overexpressing cancer cells. In conclusion, the results of our study reveal that ABCB1 negatively affects the efficacy of volasertib and supports its combination with a modulator of ABCB1 to improve clinical responses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chiun-Wei Huang
- Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital , Tao-Yuan, Taiwan
| | | |
Collapse
|
46
|
Palmisiano ND, Kasner MT. Polo-like kinase and its inhibitors: Ready for the match to start? Am J Hematol 2015; 90:1071-6. [PMID: 26294255 DOI: 10.1002/ajh.24177] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 08/06/2015] [Accepted: 08/17/2015] [Indexed: 12/25/2022]
Abstract
Polo-like kinases (Plks) plays a central role in the normal cell cycle and their upregulation has been shown to play a role in the pathogenesis of multiple human cancers. Preclinical work demonstrates that targeting Plk has a significant impact on the treatment of both solid and hematologic malignancies in vitro and in vivo. We review here the basic science and clinical work to date with the Plks as well as future directions with this novel class of mitotic inhibitors.
Collapse
|
47
|
Kobayashi Y, Yamauchi T, Kiyoi H, Sakura T, Hata T, Ando K, Watabe A, Harada A, Taube T, Miyazaki Y, Naoe T. Phase I trial of volasertib, a Polo-like kinase inhibitor, in Japanese patients with acute myeloid leukemia. Cancer Sci 2015; 106:1590-5. [PMID: 26471242 PMCID: PMC4714695 DOI: 10.1111/cas.12814] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 09/06/2015] [Indexed: 12/18/2022] Open
Abstract
This phase I trial conducted in Japanese patients with acute myeloid leukemia evaluated the safety, maximum tolerated dose and pharmacokinetics of volasertib (BI 6727), a selective Polo-like kinase inhibitor. The primary endpoints were the maximum tolerated dose of volasertib and the incidence of dose-limiting toxicities. Secondary endpoints were best response and remission duration. Other endpoints included safety and pharmacokinetics. Patients who were ineligible for standard induction therapy or with relapsed or refractory disease received volasertib monotherapy as a 2-h infusion on days 1 and 15 of a 28-day cycle, with dose escalation following a 3 + 3 design. A total of 19 patients were treated with three volasertib doses: 350, 400 and 450 mg. One patient receiving volasertib 450 mg reported a dose-limiting toxicity of grade 4 abnormal liver function test and 450 mg was determined as the maximum tolerated dose. The most frequently reported adverse events were febrile neutropenia (78.9%), decreased appetite (42.1%), nausea and rash (36.8% each), and sepsis, fatigue, hypokalemia, stomatitis and epistaxis (26.3% each). Best responses were complete remission (n = 3), complete remission with incomplete blood count recovery (n = 3) and partial remission (n = 1). The median remission duration of the six patients with complete remission or complete remission with incomplete blood count recovery was 85 days (range 56-358). Volasertib exhibited multi-compartmental pharmacokinetic behavior with a fast distribution after the end of infusion followed by slower elimination phases. Volasertib monotherapy was clinically manageable with acceptable adverse events and anti-leukemic activity.
Collapse
Affiliation(s)
- Yukio Kobayashi
- Hematology DivisionNational Cancer Center HospitalTokyoJapan
| | | | - Hitoshi Kiyoi
- Department of Hematology and OncologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Toru Sakura
- Leukemia Research CenterSaiseikai Maebashi HospitalGunmaJapan
| | - Tomoko Hata
- Department of HematologyNagasaki University HospitalNagasakiJapan
| | - Kiyoshi Ando
- Department of Hematology and OncologyTokai University School of MedicineKanagawaJapan
| | - Aiko Watabe
- Clinical Trial Management DepartmentNippon Boehringer Ingelheim Co., LtdTokyoJapan
| | - Akiko Harada
- Clinical PK/PD DepartmentNippon Boehringer Ingelheim Co., LtdHyogoJapan
| | - Tillmann Taube
- Boehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Yasushi Miyazaki
- Department of HematologyNagasaki University HospitalNagasakiJapan
| | - Tomoki Naoe
- National Hospital Organization Nagoya Medical CenterNagoyaJapan
| |
Collapse
|
48
|
Kazazian K, Brashavitskaya O, Zih FSW, Berger-Richardson D, Xu RSZ, Pacholczyk K, Macmillan J, Swallow CJ. Polo-Like Kinases in Colorectal Cancer: Potential for Targeted Therapy. CURRENT COLORECTAL CANCER REPORTS 2015. [DOI: 10.1007/s11888-015-0275-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
49
|
Park JE, Kim TS, Kim BY, Lee KS. Selective blockade of cancer cell proliferation and anchorage-independent growth by Plk1 activity-dependent suicidal inhibition of its polo-box domain. Cell Cycle 2015; 14:3624-34. [PMID: 26513691 PMCID: PMC4825759 DOI: 10.1080/15384101.2015.1104435] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 09/23/2015] [Accepted: 09/29/2015] [Indexed: 12/27/2022] Open
Abstract
Polo-like kinase 1 (Plk1) plays a critical role in proper M-phase progression and cell proliferation. Plk1 is overexpressed in a broad spectrum of human cancers and is considered an attractive anticancer drug target. Although a large number of inhibitors targeting the catalytic domain of Plk1 have been developed, these inhibitors commonly exhibit a substantial level of cross-reactivity with other structurally related kinases, thus narrowing their applicable dose for patient treatment. Plk1 contains a C-terminal polo-box domain (PBD) that is essentially required for interacting with its binding targets. However, largely due to the lack of both specific and membrane-permeable inhibitors, whether PBD serves as an alternative target for the development of anticancer therapeutics has not been rigorously examined. Here, we used an intracellularly expressed 29-mer-long PBIP1-derived peptide (i.e., PBIPtide), which can be converted into a "suicidal" PBD inhibitor via Plk1-dependent self-priming and binding. Using this highly specific and potent system, we showed that Plk1 PBD inhibition alone is sufficient for inducing mitotic arrest and apoptotic cell death in cancer cells but not in normal cells, and that cancer cell-selective killing can occur regardless of the presence or absence of oncogenic RAS mutation. Intriguingly, PBD inhibition also effectively prevented anchorage-independent growth of malignant cancer cells. Thus, targeting PBD represents an appealing strategy for anti-Plk1 inhibitor development. Additionally, PBD inhibition-induced cancer cell-selective killing may not simply stem from activated RAS alone but, rather, from multiple altered biochemical and physiological mechanisms, which may have collectively contributed to Plk1 addiction in cancer cells.
Collapse
Affiliation(s)
- Jung-Eun Park
- Laboratory of Metabolism; National Cancer Institute; National Institutes of Health; Bethesda, MD USA
| | - Tae-Sung Kim
- Laboratory of Metabolism; National Cancer Institute; National Institutes of Health; Bethesda, MD USA
| | - Bo Yeon Kim
- Incurable Diseases Therapeutics Research Center; Korea Research Institute of Bioscience and Biotechnology; Ochang, Republic of Korea
| | - Kyung S Lee
- Laboratory of Metabolism; National Cancer Institute; National Institutes of Health; Bethesda, MD USA
| |
Collapse
|