51
|
Scheiblecker L, Kollmann K, Sexl V. CDK4/6 and MAPK-Crosstalk as Opportunity for Cancer Treatment. Pharmaceuticals (Basel) 2020; 13:E418. [PMID: 33255177 PMCID: PMC7760252 DOI: 10.3390/ph13120418] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/20/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023] Open
Abstract
Despite the development of targeted therapies and novel inhibitors, cancer remains an undefeated disease. Resistance mechanisms arise quickly and alternative treatment options are urgently required, which may be partially met by drug combinations. Protein kinases as signaling switchboards are frequently deregulated in cancer and signify vulnerable nodes and potential therapeutic targets. We here focus on the cell cycle kinase CDK6 and on the MAPK pathway and on their interplay. We also provide an overview on clinical studies examining the effects of combinational treatments currently explored for several cancer types.
Collapse
Affiliation(s)
| | | | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (L.S.); (K.K.)
| |
Collapse
|
52
|
A first-in-class CDK4 inhibitor demonstrates in vitro, ex-vivo and in vivo efficacy against ovarian cancer. Gynecol Oncol 2020; 159:827-838. [PMID: 32958271 DOI: 10.1016/j.ygyno.2020.09.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/07/2020] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Cyclin-dependent kinases 4 and 6 (CDK4/6) are fundamental drivers of the cell cycle and are involved in the initiation and progression of various cancers. Deregulation of the CDK4/6-cyclin D-retinoblastoma (Rb) pathway is common in ovarian cancer and is associated with an aggressive phenotype and poor prognosis. Patients with advanced ovarian cancer whose tumor demonstrates Rb-positivity, a low expression of p16 and overexpression of cyclin D1 are most likely to benefit from CDK4/6 inhibition. MATERIALS AND METHOD Anti-proliferative activity and mechanistic investigations for CDDD2-94, employing palbociclib as comparator, were evaluated by MTT assay, cell cycle and apoptosis analysis, western blotting as well as senescence and colony formation assay. In vivo safety and efficacy studies were done in A2780 tumor-bearing nude mice. Combinations of CDDD2-94 with mTOR, MEK, PI3K or PARP inhibitors were evaluated in A2780 and OVCAR5 ovarian cancer cells. RESULTS Consistent with a CDK4-targeted mechanism, CDDD2-94 arrested the G1/G0 cell cycle, induced senescence and inhibited the proliferation of Rb-proficient ovarian cancer cells. CDDD2-94 exhibited synergistic anti-proliferative activities with mTOR, MEK, PI3K or PARP inhibitors. Importantly, unlike palbociclib which caused significant reductions in the number of lymphocytes and neutrophils, CDDD2-94 had little effect. CDDD2-94, as single agent and in combination with everolimus, delayed tumor growth and significantly increased survival of mice. CONCLUSION Given its high specificity in targeting CDK4 and excellent anti-tumor efficacy with low toxicity, CDDD2-94 has potential to be developed as a standalone agent or in combination with targeted therapeutics for the treatment of ovarian cancer.
Collapse
|
53
|
Maurer B, Brandstoetter T, Kollmann S, Sexl V, Prchal-Murphy M. Inducible deletion of CDK4 and CDK6 - deciphering CDK4/6 inhibitor effects in the hematopoietic system. Haematologica 2020; 106:2624-2632. [PMID: 32855282 PMCID: PMC8485667 DOI: 10.3324/haematol.2020.256313] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Indexed: 11/09/2022] Open
Abstract
CDK4/6 inhibitors are considered a breakthrough in cancer therapy. Currently approved for breast cancer treatment, CDK4/6 inhibitors are extensively tested in other cancer subtypes. Frequently observed side effects include hematological abnormalities such as reduced numbers of neutrophils, erythroid cells and platelets that are associated with anemia, bleedings and a higher risk of infections. To understand whether the adverse effects within the hematopoietic system are related to CDK4 or CDK6 we generated transgenic mice that lack either CDK4 or CDK6 in adult hematopoiesis. Anemia and perturbed erythroid differentiation are associated with the absence of CDK6 but did not manifest in CDK4-deficient mice. Total CDK6 knockout mice accumulate the most dormant fraction of hematopoietic stem cells due to an impaired exit of the quiescent state. We recapitulated this finding by deleting CDK6 in adult hematopoiesis. In addition, unlike total CDK6 knockout, all stem cell fractions were affected and increased in numbers. The deletion of CDK6 was also accompanied by neutropenia which is frequently seen in patients receiving CDK4/6 inhibitors. This was not the case in the absence of CDK4; CDK4 deficiency resulted in elevated numbers of myeloid progenitors without translating into numeric changes of differentiated myeloid cells. By using Cdk4 fl/fl and Cdk6 fl/fl mice we assign side effects of CDK4/6 inhibitors predominantly to the absence of CDK6. These mice represent a novel and powerful tool that will enable to study the distinct functions of CDK4 and CDK6 in a tissue-dependent manner.
Collapse
Affiliation(s)
- Barbara Maurer
- Insititute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Tania Brandstoetter
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Sebastian Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Michaela Prchal-Murphy
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
54
|
Eguchi M, Minami Y, Kuzume A, Chi S. Mechanisms Underlying Resistance to FLT3 Inhibitors in Acute Myeloid Leukemia. Biomedicines 2020; 8:biomedicines8080245. [PMID: 32722298 PMCID: PMC7459983 DOI: 10.3390/biomedicines8080245] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/10/2020] [Accepted: 07/16/2020] [Indexed: 01/03/2023] Open
Abstract
FLT3-ITD and FLT3-TKD mutations were observed in approximately 20 and 10% of acute myeloid leukemia (AML) cases, respectively. FLT3 inhibitors such as midostaurin, gilteritinib and quizartinib show excellent response rates in patients with FLT3-mutated AML, but its duration of response may not be sufficient yet. The majority of cases gain secondary resistance either by on-target and off-target abnormalities. On-target mutations (i.e., FLT3-TKD) such as D835Y keep the TK domain in its active form, abrogating pharmacodynamics of type II FLT3 inhibitors (e.g., midostaurin and quizartinib). Second generation type I inhibitors such as gilteritinib are consistently active against FLT3-TKD as well as FLT3-ITD. However, a “gatekeeper” mutation F691L shows universal resistance to all currently available FLT3 inhibitors. Off-target abnormalities are consisted with a variety of somatic mutations such as NRAS, AXL and PIM1 that bypass or reinforce FLT3 signaling. Off-target mutations can occur just in the primary FLT3-mutated clone or be gained by the evolution of other clones. A small number of cases show primary resistance by an FL-dependent, FGF2-dependent, and stromal CYP3A4-mediated manner. To overcome these mechanisms, the development of novel agents such as covalently-coupling FLT3 inhibitor FF-10101 and the investigation of combination therapy with different class agents are now ongoing. Along with novel agents, gene sequencing may improve clinical approaches by detecting additional targetable mutations and determining individual patterns of clonal evolution.
Collapse
Affiliation(s)
- Motoki Eguchi
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (M.E.); (A.K.); (S.C.)
| | - Yosuke Minami
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (M.E.); (A.K.); (S.C.)
- Correspondence: ; Tel.: +81-4-7133-1111; Fax: +81-7133-6502
| | - Ayumi Kuzume
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (M.E.); (A.K.); (S.C.)
- Division of Hematology/Oncology, Department of Internal Medicine, Kameda Medical Center, Kamogawa 296-8602, Japan
| | - SungGi Chi
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (M.E.); (A.K.); (S.C.)
| |
Collapse
|
55
|
Schmoellerl J, Barbosa IAM, Eder T, Brandstoetter T, Schmidt L, Maurer B, Troester S, Pham HTT, Sagarajit M, Ebner J, Manhart G, Aslan E, Terlecki-Zaniewicz S, Van der Veen C, Hoermann G, Duployez N, Petit A, Lapillonne H, Puissant A, Itzykson R, Moriggl R, Heuser M, Meisel R, Valent P, Sexl V, Zuber J, Grebien F. CDK6 is an essential direct target of NUP98 fusion proteins in acute myeloid leukemia. Blood 2020; 136:387-400. [PMID: 32344427 PMCID: PMC7115844 DOI: 10.1182/blood.2019003267] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 04/08/2020] [Indexed: 01/25/2023] Open
Abstract
Fusion proteins involving Nucleoporin 98 (NUP98) are recurrently found in acute myeloid leukemia (AML) and are associated with poor prognosis. Lack of mechanistic insight into NUP98-fusion-dependent oncogenic transformation has so far precluded the development of rational targeted therapies. We reasoned that different NUP98-fusion proteins deregulate a common set of transcriptional targets that might be exploitable for therapy. To decipher transcriptional programs controlled by diverse NUP98-fusion proteins, we developed mouse models for regulatable expression of NUP98/NSD1, NUP98/JARID1A, and NUP98/DDX10. By integrating chromatin occupancy profiles of NUP98-fusion proteins with transcriptome profiling upon acute fusion protein inactivation in vivo, we defined the core set of direct transcriptional targets of NUP98-fusion proteins. Among those, CDK6 was highly expressed in murine and human AML samples. Loss of CDK6 severely attenuated NUP98-fusion-driven leukemogenesis, and NUP98-fusion AML was sensitive to pharmacologic CDK6 inhibition in vitro and in vivo. These findings identify CDK6 as a conserved, critical direct target of NUP98-fusion proteins, proposing CDK4/CDK6 inhibitors as a new rational treatment option for AML patients with NUP98-fusions.
Collapse
MESH Headings
- Animals
- Cyclin-Dependent Kinase 6/antagonists & inhibitors
- Cyclin-Dependent Kinase 6/genetics
- Cyclin-Dependent Kinase 6/metabolism
- Drug Delivery Systems
- Gene Expression Profiling
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Mice
- Nuclear Pore Complex Proteins/genetics
- Nuclear Pore Complex Proteins/metabolism
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
Collapse
Affiliation(s)
- Johannes Schmoellerl
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Austria
| | | | - Thomas Eder
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Austria
| | - Tania Brandstoetter
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Austria
| | - Luisa Schmidt
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Austria
| | - Barbara Maurer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Austria
| | - Selina Troester
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Austria
| | - Ha Thi Thanh Pham
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Austria
| | - Mohanty Sagarajit
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Jessica Ebner
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Austria
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Gabriele Manhart
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Austria
| | - Ezgi Aslan
- Team of Project Machine, Medical Faculty, Istanbul Medeniyet University, Istanbul, Turkey
| | | | | | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Austria
- Central Institute for Medical and Chemical Laboratory Diagnosis, University Hospital Innsbruck, Innsbruck, Austria
| | - Nicolas Duployez
- Laboratory of Hematology, INSERM UMR-S 1172, Lille University Hospital, France
| | - Arnaud Petit
- Hopital Trousseau, Assistance Publique -Hopitaux de Paris, Paris, France
| | - Helene Lapillonne
- Hopital Trousseau, Assistance Publique -Hopitaux de Paris, Paris, France
| | - Alexandre Puissant
- INSERM U944, Saint-Louis Research Institute, University of Paris, Paris, France
| | - Raphael Itzykson
- INSERM U944, Saint-Louis Research Institute, University of Paris, Paris, France
| | - Richard Moriggl
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Austria
- Medical University of Vienna, Vienna, Austria
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Roland Meisel
- Division of Pediatric Stem Cell Therapy, Clinic for Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Austria
| | - Johannes Zuber
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
- Medical University of Vienna, Vienna, Austria
| | - Florian Grebien
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Austria
| |
Collapse
|
56
|
Voisset E, Brenet F, Lopez S, de Sepulveda P. SRC-Family Kinases in Acute Myeloid Leukaemia and Mastocytosis. Cancers (Basel) 2020; 12:cancers12071996. [PMID: 32708273 PMCID: PMC7409304 DOI: 10.3390/cancers12071996] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/17/2020] [Accepted: 07/19/2020] [Indexed: 11/16/2022] Open
Abstract
Protein tyrosine kinases have been recognized as important actors of cell transformation and cancer progression, since their discovery as products of viral oncogenes. SRC-family kinases (SFKs) play crucial roles in normal hematopoiesis. Not surprisingly, they are hyperactivated and are essential for membrane receptor downstream signaling in hematological malignancies such as acute myeloid leukemia (AML) and mastocytosis. The precise roles of SFKs are difficult to delineate due to the number of substrates, the functional redundancy among members, and the use of tools that are not selective. Yet, a large num ber of studies have accumulated evidence to support that SFKs are rational therapeutic targets in AML and mastocytosis. These two pathologies are regulated by two related receptor tyrosine kinases, which are well known in the field of hematology: FLT3 and KIT. FLT3 is one of the most frequently mutated genes in AML, while KIT oncogenic mutations occur in 80-90% of mastocytosis. Studies on oncogenic FLT3 and KIT signaling have shed light on specific roles for members of the SFK family. This review highlights the central roles of SFKs in AML and mastocytosis, and their interconnection with FLT3 and KIT oncoproteins.
Collapse
|
57
|
Zhang J, Liu X, Zhou W, Cheng G, Wu J, Guo S, Jia S, Liu Y, Li B, Zhang X, Wang M. A bioinformatics investigation into molecular mechanism of Yinzhihuang granules for treating hepatitis B by network pharmacology and molecular docking verification. Sci Rep 2020; 10:11448. [PMID: 32651427 PMCID: PMC7351787 DOI: 10.1038/s41598-020-68224-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/22/2020] [Indexed: 12/17/2022] Open
Abstract
Yinzhihuang granules (YZHG) is a patented Chinese medicine for the treatment of hepatitis B. This study aimed to investigate the intrinsic mechanisms of YZHG in the treatment of hepatitis B and to provide new evidence and insights for its clinical application. The chemical compounds of YZHG were searched in the CNKI and PUBMED databases, and their putative targets were then predicted through a search of the SuperPred and Swiss Target Prediction databases. In addition, the targets of hepatitis B were obtained from TTD, PharmGKB and DisGeNET. The abovementioned data were visualized using Cytoscape 3.7.1, and network construction identified a total of 13 potential targets of YZHG in the treatment of hepatitis B. Molecular docking verification showed that CDK6, CDK2, TP53 and BRCA1 might be strongly correlated with hepatitis B treatment. Furthermore, GO and KEGG analyses indicated that the treatment of hepatitis B by YZHG might be related to positive regulation of transcription, positive regulation of gene expression, the hepatitis B pathway and the viral carcinogenesis pathway. Network pharmacology intuitively shows the multicomponent, multitarget and multichannel pharmacological effects of YZHG in the treatment of hepatitis B and provides a scientific basis for its mechanism of action.
Collapse
Affiliation(s)
- Jingyuan Zhang
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Xinkui Liu
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Wei Zhou
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Guoliang Cheng
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, 276000, China
| | - Jiarui Wu
- Beijing University of Chinese Medicine, Beijing, 100102, China.
| | - Siyu Guo
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Shanshan Jia
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Yingying Liu
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Bingbing Li
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, 276000, China
| | - Xiaomeng Zhang
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Miaomiao Wang
- Beijing University of Chinese Medicine, Beijing, 100102, China
| |
Collapse
|
58
|
Yamashita M, Dellorusso PV, Olson OC, Passegué E. Dysregulated haematopoietic stem cell behaviour in myeloid leukaemogenesis. Nat Rev Cancer 2020; 20:365-382. [PMID: 32415283 PMCID: PMC7658795 DOI: 10.1038/s41568-020-0260-3] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/02/2020] [Indexed: 12/17/2022]
Abstract
Haematopoiesis is governed by haematopoietic stem cells (HSCs) that produce all lineages of blood and immune cells. The maintenance of blood homeostasis requires a dynamic response of HSCs to stress, and dysregulation of these adaptive-response mechanisms underlies the development of myeloid leukaemia. Leukaemogenesis often occurs in a stepwise manner, with genetic and epigenetic changes accumulating in pre-leukaemic HSCs prior to the emergence of leukaemic stem cells (LSCs) and the development of acute myeloid leukaemia. Clinical data have revealed the existence of age-related clonal haematopoiesis, or the asymptomatic clonal expansion of mutated blood cells in the elderly, and this phenomenon is connected to susceptibility to leukaemic transformation. Here we describe how selection for specific mutations that increase HSC competitive fitness, in conjunction with additional endogenous and environmental changes, drives leukaemic transformation. We review the ways in which LSCs take advantage of normal HSC properties to promote survival and expansion, thus underlying disease recurrence and resistance to conventional therapies, and we detail our current understanding of leukaemic 'stemness' regulation. Overall, we link the cellular and molecular mechanisms regulating HSC behaviour with the functional dysregulation of these mechanisms in myeloid leukaemia and discuss opportunities for targeting LSC-specific mechanisms for the prevention or cure of malignant diseases.
Collapse
Affiliation(s)
- Masayuki Yamashita
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Paul V Dellorusso
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Oakley C Olson
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
59
|
Inhibition of Rb phosphorylation leads to H 2S-mediated inhibition of NF-kB in acute pancreatitis and associated lung injury in mice. Pancreatology 2020; 20:647-658. [PMID: 32402695 DOI: 10.1016/j.pan.2020.04.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 04/04/2020] [Accepted: 04/16/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Acute pancreatitis (AP), an inflammatory condition of pancreas, destructs the exocrine cells by releasing various pro-inflammatory cytokines that activates the stellate cells. However, the underlying molecular mechanism remains unclear. The present study investigated the role of retinoblastoma (Rb), hydrogen sulphide and nuclear factor-κB (NF-κB) in the regulation of exocrine cell proliferation under inflammatory condition. METHODS The randomly grouped male swiss mice were administered with 6 consecutive hourly i.p injections of caerulein to induce AP. Palbociclib (PD) (25 mg/kg body weight), a CDK4/6 inhibitor, was administered 1 h after the first cerulein injection intraperitoneally to block the RB pathway by inhibiting the activity of the CDK4/6 complexes and DL propargylglycine (PAG) which blocks the endogenous H2S production. RESULTS Pharmacological inhibition of CDK4/6 and H2S significantly improved pancreas and lung histopathological changes, decreased serum amylase level, both lung and pancreas myeloperoxidase (MPO) activity, TNFα expression and elevated IL10 expression. Furthermore, inhibition of RB pathway reduced cerulein-induced H2S level by reducing the expression of cystathionine gamma lyase (CSE) and NF-κB activation in pancreas and lungs. Also, blocking the RB signalling reduced the α-SMA expression in pancreas preventing the risk for pancreatic fibrosis. Whereas administration of H2S inhibitor PAG resulted in a decrease in CDK4/6-Rb expression in cerulein-induced AP. CONCLUSION These results reveal a novel link between H2S/RB/NF-κB pathways, in AP and provide insight into possible mechanism that can be targeted in prevention of inflammation to cancer development.
Collapse
|
60
|
Nebenfuehr S, Kollmann K, Sexl V. The role of CDK6 in cancer. Int J Cancer 2020; 147:2988-2995. [PMID: 32406095 PMCID: PMC7586846 DOI: 10.1002/ijc.33054] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/29/2020] [Accepted: 05/04/2020] [Indexed: 12/27/2022]
Abstract
The regulation and function of cyclin‐dependent kinase 6 (CDK6)‐ and cyclin‐dependent kinase 4 (CDK4)‐cyclin complexes are commonly altered with enhanced kinase activity found in hematopoietic malignancies, breast cancer and melanoma making CDK4 and CDK6 attractive targets for therapeutic interference. Although dual CDK4/6 inhibitors have revolutionized treatment of breast cancer patients and reveal promising results in several solid tumors and hematological malignancies, there is a need for novel compounds targeting the versatile kinase‐independent functions of CDK6 to improve cancer treatment. The following review summarizes the latest findings on CDK6 in cancer development and discusses novel therapeutic approaches to selectively inhibit CDK6s function as a transcriptional regulator.
Collapse
Affiliation(s)
- Sofie Nebenfuehr
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Karoline Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
61
|
Uras IZ, Sexl V, Kollmann K. CDK6 Inhibition: A Novel Approach in AML Management. Int J Mol Sci 2020; 21:ijms21072528. [PMID: 32260549 PMCID: PMC7178035 DOI: 10.3390/ijms21072528] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 03/29/2020] [Accepted: 04/02/2020] [Indexed: 02/01/2023] Open
Abstract
Acute myeloid leukemia (AML) is a complex disease with an aggressive clinical course and high mortality rate. The standard of care for patients has only changed minimally over the past 40 years. However, potentially useful agents have moved from bench to bedside with the potential to revolutionize therapeutic strategies. As such, cell-cycle inhibitors have been discussed as alternative treatment options for AML. In this review, we focus on cyclin-dependent kinase 6 (CDK6) emerging as a key molecule with distinct functions in different subsets of AML. CDK6 exerts its effects in a kinase-dependent and -independent manner which is of clinical significance as current inhibitors only target the enzymatic activity.
Collapse
Affiliation(s)
- Iris Z. Uras
- Department of Pharmacology, Center of Physiology and Pharmacology & Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria;
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Karoline Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria;
- Correspondence: ; Tel.: + 43-1-25077-2917
| |
Collapse
|
62
|
circMYBL2, a circRNA from MYBL2, regulates FLT3 translation by recruiting PTBP1 to promote FLT3-ITD AML progression. Blood 2020; 134:1533-1546. [PMID: 31387917 DOI: 10.1182/blood.2019000802] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 08/03/2019] [Indexed: 01/16/2023] Open
Abstract
Internal tandem duplication (ITD) mutations within FMS-like tyrosine kinase-3 (FLT3) occur in up to 30% of acute myeloid leukemia (AML) patients and confer a very poor prognosis. The oncogenic form of FLT3 is an important therapeutic target, and inhibitors specifically targeting FLT3 kinase can induce complete remission; however, relapse after remission has been observed due to acquired resistance with secondary mutations in FLT3, highlighting the need for new strategies to target FLT3-ITD mutations. Recent studies have reported that the aberrant formations of circular RNAs (circRNAs) are biological tumorigenesis-relevant mechanisms and potential therapeutic targets. Herein, we discovered a circRNA, circMYBL2, derived from the cell-cycle checkpoint gene MYBL2. circMYBL2 is more highly expressed in AML patients with FLT3-ITD mutations than in those without the FLT3-ITD mutation. We found that circMYBL2 knockdown specifically inhibits proliferation and promotes the differentiation of FLT3-ITD AML cells in vitro and in vivo. Interestingly, we found that circMYBL2 significantly influences the protein level of mutant FLT3 kinase, which contributes to the activation of FLT3-ITD-dependent signaling pathways. Mechanistically, circMYBL2 enhanced the translational efficiency of FLT3 kinase by increasing the binding of polypyrimidine tract-binding protein 1 (PTBP1) to FLT3 messenger RNA. Moreover, circMYBL2 knockdown impaired the cytoactivity of inhibitor-resistant FLT3-ITD+ cells, with a significant decrease in FLT3 kinase expression, followed by the inactivation of its downstream pathways. In summary, we are the first to reveal a circRNA that specifically influences FLT3-ITD AML and regulates FLT3 kinase levels through translational regulation, suggesting that circMYBL2 may be a potential therapeutic target for FLT3-ITD AML.
Collapse
|
63
|
Parra M, Baptista MJ, Genescà E, Llinàs-Arias P, Esteller M. Genetics and epigenetics of leukemia and lymphoma: from knowledge to applications, meeting report of the Josep Carreras Leukaemia Research Institute. Hematol Oncol 2020; 38:432-438. [PMID: 32073154 PMCID: PMC7687178 DOI: 10.1002/hon.2725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 02/08/2020] [Indexed: 12/21/2022]
Abstract
The meeting, which brought together leading scientists and clinicians in the field of leukemia and lymphoma, was held at the new headquarters of the Josep Carreras Leukaemia Research Institute (IJC) in Badalona, Catalonia, Spain, September 19-20, 2019. Its purpose was to highlight the latest advances in our understanding of the molecular mechanisms driving blood cancers, and to discuss how this knowledge can be translated into an improved management of the disease. Special emphasis was placed on the role of genetic and epigenetic heterogeneity, and the exploitation of epigenetic regulation for developing biomarkers and novel treatment approaches.
Collapse
Affiliation(s)
- Maribel Parra
- Lymphocyte Development and Disease Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Maria Joao Baptista
- Lymphoid neoplasms Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Eulàlia Genescà
- Acute lymphoblastic leukemia (ALL) Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Pere Llinàs-Arias
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain.,Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain.,Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.,Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain
| |
Collapse
|
64
|
Short NJ, Konopleva M, Kadia TM, Borthakur G, Ravandi F, DiNardo CD, Daver N. Advances in the Treatment of Acute Myeloid Leukemia: New Drugs and New Challenges. Cancer Discov 2020; 10:506-525. [DOI: 10.1158/2159-8290.cd-19-1011] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/23/2019] [Accepted: 11/20/2019] [Indexed: 11/16/2022]
|
65
|
Chen X, Burkhardt DB, Hartman AA, Hu X, Eastman AE, Sun C, Wang X, Zhong M, Krishnaswamy S, Guo S. MLL-AF9 initiates transformation from fast-proliferating myeloid progenitors. Nat Commun 2019; 10:5767. [PMID: 31852898 PMCID: PMC6920141 DOI: 10.1038/s41467-019-13666-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 11/19/2019] [Indexed: 01/16/2023] Open
Abstract
Cancer is a hyper-proliferative disease. Whether the proliferative state originates from the cell-of-origin or emerges later remains difficult to resolve. By tracking de novo transformation from normal hematopoietic progenitors expressing an acute myeloid leukemia (AML) oncogene MLL-AF9, we reveal that the cell cycle rate heterogeneity among granulocyte-macrophage progenitors (GMPs) determines their probability of transformation. A fast cell cycle intrinsic to these progenitors provide permissiveness for transformation, with the fastest cycling 3% GMPs acquiring malignancy with near certainty. Molecularly, we propose that MLL-AF9 preserves gene expression of the cellular states in which it is expressed. As such, when expressed in the naturally-existing, rapidly-cycling immature myeloid progenitors, this cell state becomes perpetuated, yielding malignancy. In humans, high CCND1 expression predicts worse prognosis for MLL fusion AMLs. Our work elucidates one of the earliest steps toward malignancy and suggests that modifying the cycling state of the cell-of-origin could be a preventative approach against malignancy.
Collapse
MESH Headings
- Animals
- Cell Cycle/drug effects
- Cell Cycle/genetics
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Cell Proliferation/drug effects
- Cell Proliferation/genetics
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/genetics
- Cyclin D1/metabolism
- Disease Models, Animal
- Female
- Gene Expression Regulation, Leukemic
- Gene Knock-In Techniques
- Humans
- Kaplan-Meier Estimate
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Male
- Mice, Transgenic
- Myeloid Progenitor Cells/pathology
- Myeloid-Lymphoid Leukemia Protein/genetics
- Oncogene Proteins, Fusion/genetics
- Piperazines/administration & dosage
- Primary Cell Culture
- Prognosis
- Pyridines/administration & dosage
Collapse
Affiliation(s)
- Xinyue Chen
- Department of Cell Biology, Yale University, New Haven, CT 06520 USA
- Yale Stem Cell Center, Yale University, New Haven, CT 06520 USA
| | | | - Amaleah A. Hartman
- Department of Cell Biology, Yale University, New Haven, CT 06520 USA
- Yale Stem Cell Center, Yale University, New Haven, CT 06520 USA
| | - Xiao Hu
- Department of Cell Biology, Yale University, New Haven, CT 06520 USA
- Yale Stem Cell Center, Yale University, New Haven, CT 06520 USA
| | - Anna E. Eastman
- Department of Cell Biology, Yale University, New Haven, CT 06520 USA
- Yale Stem Cell Center, Yale University, New Haven, CT 06520 USA
| | - Chao Sun
- Department of Cell Biology, Yale University, New Haven, CT 06520 USA
- Yale Stem Cell Center, Yale University, New Haven, CT 06520 USA
| | - Xujun Wang
- SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, 200240 China
| | - Mei Zhong
- Department of Cell Biology, Yale University, New Haven, CT 06520 USA
- Yale Stem Cell Center, Yale University, New Haven, CT 06520 USA
| | | | - Shangqin Guo
- Department of Cell Biology, Yale University, New Haven, CT 06520 USA
- Yale Stem Cell Center, Yale University, New Haven, CT 06520 USA
| |
Collapse
|
66
|
Yang T, Hu M, Qi W, Yang Z, Tang M, He J, Chen Y, Bai P, Yuan X, Zhang C, Liu K, Lu Y, Xiang M, Chen L. Discovery of Potent and Orally Effective Dual Janus Kinase 2/FLT3 Inhibitors for the Treatment of Acute Myelogenous Leukemia and Myeloproliferative Neoplasms. J Med Chem 2019; 62:10305-10320. [PMID: 31670517 DOI: 10.1021/acs.jmedchem.9b01348] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Tao Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Mengshi Hu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Wenyan Qi
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhuang Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Minghai Tang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Jun He
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Yong Chen
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Peng Bai
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Xue Yuan
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Chufeng Zhang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Kongjun Liu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Yulin Lu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Mingli Xiang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
67
|
Lee DJ, Zeidner JF. Cyclin-dependent kinase (CDK) 9 and 4/6 inhibitors in acute myeloid leukemia (AML): a promising therapeutic approach. Expert Opin Investig Drugs 2019; 28:989-1001. [PMID: 31612739 DOI: 10.1080/13543784.2019.1678583] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Despite advancements over the last 2 years, outcomes for acute myeloid leukemia (AML) are poor; however, a greater comprehension of disease mechanisms has driven the investigation of new targeted treatments. Cyclin-dependent kinases (CDKs) regulate cell cycle progression, transcription and DNA repair, and are aberrantly expressed in AML. Targeting the CDK pathway is an emerging promising therapeutic strategy in AML.Areas covered: We describe the rationale for targeting CDK9 and CDK4/6, the ongoing preclinical and clinical trials and the potential of these inhibitors in AML. Our analysis included an extensive literature search via the Pubmed database and clinicaltrials.gov (March to August, 2019).Expert opinion: While CDK4/6 inhibitors are early in development for AML, CDK9 inhibition with alvocidib has encouraging clinical activity in newly diagnosed and relapsed/refractory AML. Preclinical data suggests that leukemic MCL-1 dependence may predict response to alvocidib. Moreover, MCL-1 plays a key role in resistance to BCL-2 inhibition with venetoclax. Investigational strategies of concomitant BCL-2 and CDK9 inhibition represent a promising therapeutic platform for AML. Furthermore, preclinical data suggests that CDK4/6 inhibition has selective activity in patients with KMT2A-rearrangements and FLT3 mutations. Incorporation of CDK9 and 4/6 inhibitors into the existing therapeutic armamentarium may improve outcomes in AML.
Collapse
Affiliation(s)
- Daniel J Lee
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, USA
| | - Joshua F Zeidner
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina, Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| |
Collapse
|
68
|
Menzl I, Zhang T, Berger-Becvar A, Grausenburger R, Heller G, Prchal-Murphy M, Edlinger L, Knab VM, Uras IZ, Grundschober E, Bauer K, Roth M, Skucha A, Liu Y, Hatcher JM, Liang Y, Kwiatkowski NP, Fux D, Hoelbl-Kovacic A, Kubicek S, Melo JV, Valent P, Weichhart T, Grebien F, Zuber J, Gray NS, Sexl V. A kinase-independent role for CDK8 in BCR-ABL1 + leukemia. Nat Commun 2019; 10:4741. [PMID: 31628323 PMCID: PMC6802219 DOI: 10.1038/s41467-019-12656-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 09/20/2019] [Indexed: 12/19/2022] Open
Abstract
Cyclin-dependent kinases (CDKs) are frequently deregulated in cancer and represent promising drug targets. We provide evidence that CDK8 has a key role in B-ALL. Loss of CDK8 in leukemia mouse models significantly enhances disease latency and prevents disease maintenance. Loss of CDK8 is associated with pronounced transcriptional changes, whereas inhibiting CDK8 kinase activity has minimal effects. Gene set enrichment analysis suggests that the mTOR signaling pathway is deregulated in CDK8-deficient cells and, accordingly, these cells are highly sensitive to mTOR inhibitors. Analysis of large cohorts of human ALL and AML patients reveals a significant correlation between the level of CDK8 and of mTOR pathway members. We have synthesized a small molecule YKL-06-101 that combines mTOR inhibition and degradation of CDK8, and induces cell death in human leukemic cells. We propose that simultaneous CDK8 degradation and mTOR inhibition might represent a potential therapeutic strategy for the treatment of ALL patients. Cyclin-dependent kinases are deregulated in blood cancers. Here, the authors show that CDK8, independent of its kinase activity, regulates mTOR signalling for the maintenance of BCR-ABL1+ leukemia, and that the dual inhibition of CDK8 and mTOR signalling induces apoptosis in these cells.
Collapse
Affiliation(s)
- Ingeborg Menzl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Tinghu Zhang
- Department of Cancer Biology, Department of Biological Chemistry and Molecular Pharmacology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Angelika Berger-Becvar
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Reinhard Grausenburger
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Gerwin Heller
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria.,Department of Medicine I, Medical University of Vienna, Vienna, Austria.,Comprehensive Cancer Center, Vienna, Austria
| | - Michaela Prchal-Murphy
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Leo Edlinger
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Vanessa M Knab
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Iris Z Uras
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Eva Grundschober
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Karin Bauer
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Mareike Roth
- Research Institute of Molecular Pathology, Campus-Vienna-Biocenter 1, Vienna, Austria
| | - Anna Skucha
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Yao Liu
- Department of Cancer Biology, Department of Biological Chemistry and Molecular Pharmacology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - John M Hatcher
- Department of Cancer Biology, Department of Biological Chemistry and Molecular Pharmacology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Yanke Liang
- Department of Cancer Biology, Department of Biological Chemistry and Molecular Pharmacology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Nicholas P Kwiatkowski
- Department of Cancer Biology, Department of Biological Chemistry and Molecular Pharmacology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniela Fux
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Andrea Hoelbl-Kovacic
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Stefan Kubicek
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Junia V Melo
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, 5005, Australia.,Department of Hematology, Imperial College London, Kensington, London, SW7 2AZ, UK
| | - Peter Valent
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Thomas Weichhart
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Florian Grebien
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Institute for Medical Biochemistry, University of Veterinary Medicine, Vienna, Austria
| | - Johannes Zuber
- Research Institute of Molecular Pathology, Campus-Vienna-Biocenter 1, Vienna, Austria
| | - Nathanael S Gray
- Department of Cancer Biology, Department of Biological Chemistry and Molecular Pharmacology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria.
| |
Collapse
|
69
|
Kazi JU, Rönnstrand L. FMS-like Tyrosine Kinase 3/FLT3: From Basic Science to Clinical Implications. Physiol Rev 2019; 99:1433-1466. [PMID: 31066629 DOI: 10.1152/physrev.00029.2018] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) is a receptor tyrosine kinase that is expressed almost exclusively in the hematopoietic compartment. Its ligand, FLT3 ligand (FL), induces dimerization and activation of its intrinsic tyrosine kinase activity. Activation of FLT3 leads to its autophosphorylation and initiation of several signal transduction cascades. Signaling is initiated by the recruitment of signal transduction molecules to activated FLT3 through binding to specific phosphorylated tyrosine residues in the intracellular region of FLT3. Activation of FLT3 mediates cell survival, cell proliferation, and differentiation of hematopoietic progenitor cells. It acts in synergy with several other cytokines to promote its biological effects. Deregulated FLT3 activity has been implicated in several diseases, most prominently in acute myeloid leukemia where around one-third of patients carry an activating mutant of FLT3 which drives the disease and is correlated with poor prognosis. Overactivity of FLT3 has also been implicated in autoimmune diseases, such as rheumatoid arthritis. The observation that gain-of-function mutations of FLT3 can promote leukemogenesis has stimulated the development of inhibitors that target this receptor. Many of these are in clinical trials, and some have been approved for clinical use. However, problems with acquired resistance to these inhibitors are common and, furthermore, only a fraction of patients respond to these selective treatments. This review provides a summary of our current knowledge regarding structural and functional aspects of FLT3 signaling, both under normal and pathological conditions, and discusses challenges for the future regarding the use of targeted inhibition of these pathways for the treatment of patients.
Collapse
Affiliation(s)
- 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 ; and Division of Oncology, Skåne University Hospital , Lund , Sweden
| | - Lars Rönnstrand
- 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 ; and Division of Oncology, Skåne University Hospital , Lund , Sweden
| |
Collapse
|
70
|
Winer ES, Stone RM. Novel therapy in Acute myeloid leukemia (AML): moving toward targeted approaches. Ther Adv Hematol 2019; 10:2040620719860645. [PMID: 31321011 PMCID: PMC6624910 DOI: 10.1177/2040620719860645] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/30/2019] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogenous and complex disease characterized by rapid cellular proliferation, an aggressive clinical course, and generally high mortality. While progress has been made in the understanding of the genetic and molecular biology of the disease, the standard of care for patients had only changed minimally over the past 40 years. Recently, rapid movement of potentially useful agents from bench to bedside has translated into new therapies either recently approved or in clinical trials. These therapies include improved chemotherapies, mutationally targeted inhibitors, pro-apoptotic agents, microenvironment targeting molecules, cell cycle checkpoint inhibitors, and epigenetic regulators. Furthermore, advances in immunotherapy employ monoclonal and bispecific antibodies, chimeric antigen receptor (CAR) T cells, checkpoint inhibitors, and vaccines provide an alternative pathway for AML treatment. In this review, we discuss the recent results of completed or ongoing clinical trials with these novel therapeutic agents in AML.
Collapse
Affiliation(s)
- Eric S. Winer
- Dana-Farber Cancer Institute, Leukemia Division,
Department of Medical Oncology, Boston, MA, USA
| | - Richard M. Stone
- Dana-Farber Cancer Institute, Leukemia Division,
Department of Medical Oncology, 450 Brookline Ave., Boston, MA 02115,
USA
| |
Collapse
|
71
|
Tallis E, Borthakur G. Novel treatments for relapsed/refractory acute myeloid leukemia with FLT3 mutations. Expert Rev Hematol 2019; 12:621-640. [PMID: 31232619 DOI: 10.1080/17474086.2019.1635882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Introduction: Mutations in the gene encoding for the FMS-like tyrosine kinase 3 (FLT3) are present in about 30% of adults with AML and are associated with shorter disease-free and overall survival after initial therapy. Prognosis of relapsed/refractory AML with FLT3 mutations is even more dismal with median overall survival of a few months only. Areas covered: This review will cover current and emerging treatments for relapsed/refractory AML with FLT3 mutations, preclinical rationale and clinical trials with new encouraging data for this particularly challenging population. The authors discuss mechanisms of resistance to FLT3 inhibitors and how these insights serve to identify current and future treatments. As allogeneic stem cell transplant in the first remission is the preferred therapy for newly diagnosed AML patients with FLT3 mutations, the authors discuss the role of maintenance after SCT for the prevention of relapse. Expert opinion: Relapsed/refractory AML with FLT3 mutations remains a therapeutic challenge with currently available treatments. However, the evolution of targeted therapies with next-generation FLT3 inhibitors and their combinations with chemotherapy is showing much promise. Moreover, growing understanding of the pathways of resistance to treatment has led to the identification of various targeted therapies currently being explored, which in time will improve outcomes.
Collapse
Affiliation(s)
- Eran Tallis
- a Department of Leukemia, The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Gautam Borthakur
- a Department of Leukemia, The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
72
|
Poratti M, Marzaro G. Third-generation CDK inhibitors: A review on the synthesis and binding modes of Palbociclib, Ribociclib and Abemaciclib. Eur J Med Chem 2019; 172:143-153. [DOI: 10.1016/j.ejmech.2019.03.064] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/18/2019] [Accepted: 03/31/2019] [Indexed: 12/20/2022]
|
73
|
Aierken K, Dong Z, Abulimiti T, Zhang Y, Abuduxikuer G, Tuerxun G, Abudurexiti G, Maimaitiaishan A, Mijiti P, Abulizi G. CDK6 3'UTR polymorphisms alter the susceptibility to cervical cancer among Uyghur females. Mol Genet Genomic Med 2019; 7:e626. [PMID: 30829464 PMCID: PMC6503018 DOI: 10.1002/mgg3.626] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/11/2019] [Accepted: 02/11/2019] [Indexed: 12/24/2022] Open
Abstract
AIMS Cyclin dependent kinase 6 (CDK6) plays a crucial role in malignant tumor whereas less is reported in cervical cancer development. The aim of this study was to evaluate the effects of CDK6 3' untranslated region (3'UTR) polymorphisms on cervical cancer susceptibility among Uyghur females. METHODS The genotypes of the six CDK6 variants (rs8179, rs42032, rs42033, rs42034, rs42035, and rs42038) were identified among 306 cervical cancer cases and 310 healthy controls with the Agena MassARRAY platform. The associations of the candidate single nucleotide polymorphisms (SNPs) with the cervical cancer risk were evaluated under genetic models using conditional logistic regression analysis. Bioinformatics analysis was performed for SNP function prediction with the online databases. The expression differences between tumor tissues and normal cervix samples were also examined by Real-time PCR. RESULTS CDK6 rs8179 and rs42033 were correlated to the decreased risk of cervical cancer in Uyghurs under the allele model (rs8179 and rs42033: OR = 0.60, 95% CI: 0.37-0.99, p = 0.043) and log-additive model (rs8179 and rs42033: OR = 0.62, 95% CI: 0.38-1.00, p = 0.047). Rs8179, rs42032, and rs42033 were associated with susceptibility to high-grade cervical cancer in different genetic models as well (p < 0.05). Dataset-based analysis also uncovered the potential effects of these significant SNPs. In addition, aberrant expression of CDK6 were detected in cervical tumors. CONCLUSIONS Our results suggested the relationships between CDK6 3'UTR polymorphisms and cervical cancer pathogenesis, and the involvement of CDK6 in cervical cancer development among Uyghur females.
Collapse
Affiliation(s)
- Kailibinuer Aierken
- 5th Department of GynecologyAffiliated Tumor Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Zhihong Dong
- Outpatient DepartmentAffiliated Tumor Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Tangnuer Abulimiti
- 5th Department of GynecologyAffiliated Tumor Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Yuanyuan Zhang
- 5th Department of GynecologyAffiliated Tumor Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Guzhalinuer Abuduxikuer
- 5th Department of GynecologyAffiliated Tumor Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Gulixian Tuerxun
- 5th Department of GynecologyAffiliated Tumor Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Guligeina Abudurexiti
- 5th Department of GynecologyAffiliated Tumor Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Aziguli Maimaitiaishan
- 5th Department of GynecologyAffiliated Tumor Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Patiman Mijiti
- 5th Department of GynecologyAffiliated Tumor Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Guzhalinuer Abulizi
- 5th Department of GynecologyAffiliated Tumor Hospital of Xinjiang Medical UniversityUrumqiChina
| |
Collapse
|
74
|
Short NJ, Kantarjian H, Ravandi F, Daver N. Emerging treatment paradigms with FLT3 inhibitors in acute myeloid leukemia. Ther Adv Hematol 2019; 10:2040620719827310. [PMID: 30800259 PMCID: PMC6378516 DOI: 10.1177/2040620719827310] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/07/2019] [Indexed: 11/17/2022] Open
Abstract
Mutations in the fms-like tyrosine kinase 3 (FLT3) gene are detected in approximately one-third of patients with newly diagnosed acute myeloid leukemia (AML). These consist of the more common FLT3-internal tandem duplication (ITD) in approximately 20-25% of AML cases, and point mutations in the tyrosine kinase domain (TKD) in approximately 5-10%. FLT3 mutations, especially FLT3-ITD, are associated with proliferative disease, increased risk of relapse, and inferior overall survival when treated with conventional regimens. However, the recent development of well tolerated and active FLT3 inhibitors has significantly improved the outcomes of this aggressive subtype of AML. The multikinase inhibitor midostaurin was approved by the United States Food and Drug Administration (US FDA) in April 2017 for the frontline treatment of patients with FLT3-mutated (either ITD or TKD) AML in combination with induction chemotherapy, representing the first new drug approval in AML in nearly two decades. In November 2018, the US FDA also approved the second-generation FLT3 inhibitor gilteritinib as a single agent for patients with relapsed or refractory FLT3-mutated AML. Promising phase I and II efficacy data for quizartinib is likely to lead to a third regulatory approval in relapsed/refractory AML in the near future. However, despite the significant progress made in managing FLT3-mutated AML, many questions remain regarding the best approach to integrate these inhibitors into combination regimens, and also the optimal sequencing of different FLT3 inhibitors in various clinical settings. This review comprehensively examines the FLT3 inhibitors currently in clinical development, with an emphasis on their spectra of activity against different FLT3 mutations and other kinases, clinical safety and efficacy data, and their current and future roles in the management of AML. The mechanisms of resistance to FLT3 inhibitors and potential combination strategies to overcome such resistance pathways are also discussed.
Collapse
Affiliation(s)
- Nicholas J. Short
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop Kantarjian
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| |
Collapse
|
75
|
Kollmann K, Briand C, Bellutti F, Schicher N, Blunder S, Zojer M, Hoeller C. The interplay of CDK4 and CDK6 in melanoma. Oncotarget 2019; 10:1346-1359. [PMID: 30858922 PMCID: PMC6402717 DOI: 10.18632/oncotarget.26515] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/04/2018] [Indexed: 11/25/2022] Open
Abstract
The cyclin-dependent kinases CDK4 and CDK6 promote progression through the cell cycle, where their functions are considered to be redundant. Recent studies have identified an additional role for CDK6 in the transcriptional regulation of cancer-relevant genes such as VEGF-A and EGR1 in hematopoietic malignancies. We show that the CDK4/6 inhibitor PD0332991 causes a significant decrease in tumor growth in a xenotransplantation mouse model of human melanoma. shRNA knockdown of either CDK4 or CDK6 significantly reduces cell proliferation and impedes their migratory capacity in vitro, which translates into a strong inhibition of tumor growth in xenotransplantation experiments. CDK4/6 inhibition results not only in the pronounced reduction of cell proliferation but also in an impaired tumor angiogenesis. CDK6 knockdown in melanoma cell lines impairs VEGF-A expression and reduces the potential stimulation of endothelial cell growth. The knockdown of CDK4 ends in similar results. The effect is caused by changes of CDK6 localization, less CDK6 is detected on the VEGF-A promoter. Bioinformatic analysis of human melanoma patient data verifies the key role of CDK6 in tumor angiogenesis in melanoma. The results highlight the importance of the delicate balance between CDK4 and CDK6 in regulating the cell cycle and transcription.
Collapse
Affiliation(s)
- Karoline Kollmann
- Institute of Pharmacology and Toxicology, Veterinary University of Vienna, Vienna, Austria
| | - Coralie Briand
- Department of Dermatology, Division of General Dermatology, Medical University Vienna, Vienna, Austria
| | - Florian Bellutti
- Institute of Pharmacology and Toxicology, Veterinary University of Vienna, Vienna, Austria
| | - Nikolaus Schicher
- Department of Dermatology, Division of General Dermatology, Medical University Vienna, Vienna, Austria
| | - Stefan Blunder
- Department of Dermatology, Division of General Dermatology, Medical University Vienna, Vienna, Austria
| | - Markus Zojer
- Institute of Pharmacology and Toxicology, Veterinary University of Vienna, Vienna, Austria
| | - Christoph Hoeller
- Department of Dermatology, Division of General Dermatology, Medical University Vienna, Vienna, Austria
| |
Collapse
|
76
|
Uscanga-Perales GI, Santuario-Facio SK, Sanchez-Dominguez CN, Cardona-Huerta S, Muñoz-Maldonado GE, Ruiz-Flores P, Barcenas-Walls JR, Osuna-Rosales LE, Rojas-Martinez A, Gonzalez-Guerrero JF, Valero-Gomez J, Gomez-Macias GS, Barbosa-Quintana A, Barboza-Quintana O, Garza-Guajardo R, Ortiz-Lopez R. Genetic alterations of triple negative breast cancer (TNBC) in women from Northeastern Mexico. Oncol Lett 2019; 17:3581-3588. [PMID: 30867801 DOI: 10.3892/ol.2019.9984] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/30/2018] [Indexed: 12/24/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a subtype of breast cancer of heterogeneous nature that is negative for estrogen receptor (ER), progesterone receptor (PR) and growth factor human epidermal 2 (HER2) following immunohistochemical analysis. TNBC is frequently characterized by relapse and reduced survival. To date, there is no targeted therapy for this type of cancer. Chemotherapy, radiotherapy, and surgery remain as the standard treatments options. The lack of a target therapy and the heterogeneity of TNBC highlight the need to seek new therapeutic options. In this study, fresh tissue samples of TNBC were analyzed with a panel of 48 driver genes (212 amplicons) that are likely to be therapeutic targets. We found intron variants, missense, stop gained and splicing variants in TP53, PIK3CA and FLT3 genes. Interestingly, all the analyzed samples had at least two variants in the TP53 gene, one being a drug response variant, rs1042522, found in 94% of our samples. We also found seven additional variants not previously reported in the TP53 gene, to the best of our knowledge, with probable deleterious characteristics of the tumor suppressor gene. We found four genetic variants in the PIK3CA gene, including two missense variants. The rs2491231 variant in the FLT3 gene was identified in 84% (16/19) of the samples, which not yet reported for TNBC, to the best of our knowledge. In conclusion, genetic variants in TP53 were found in all TNBC tumors, with rs1042522 being the most frequent (94% of TNBC biopsies), which had not been previously reported in TNBC. Also, we found two missense variants in the PIK3CA gene. These results justify the validation of these genetic variants in a large cohort, as well as the extensive study of their impact on the prognosis and therapy management of TBNC.
Collapse
Affiliation(s)
- Grecia I Uscanga-Perales
- Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autonoma de Nuevo Leon, Monterrey, Nuevo Leon 66460, Mexico.,Departamento de Bioquimica y Medicina Molecular, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Monterrey, Nuevo Leon 66460, Mexico
| | - Sandra K Santuario-Facio
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey, Nuevo Leon 64710, Mexico
| | - Celia N Sanchez-Dominguez
- Departamento de Bioquimica y Medicina Molecular, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Monterrey, Nuevo Leon 66460, Mexico
| | - Servando Cardona-Huerta
- Centro de Cancer de Mama, Hospital San Jose, Tecnologico de Monterrey, Monterrey, Nuevo Leon 64710, Mexico
| | - Gerardo E Muñoz-Maldonado
- Servicio de Cirugia General, Hospital Universitario Dr. Jose Eleuterio Gonzalez, Universidad Autonoma de Nuevo Leon, Monterrey, Nuevo Leon 66460, Mexico
| | - Pablo Ruiz-Flores
- Departamento de Medicina Molecular, Facultad de Medicina, Universidad Autonoma de Coahuila, Torreon, Coahuila 27000, Mexico
| | - Jose R Barcenas-Walls
- Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autonoma de Nuevo Leon, Monterrey, Nuevo Leon 66460, Mexico
| | - Luis E Osuna-Rosales
- Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autonoma de Nuevo Leon, Monterrey, Nuevo Leon 66460, Mexico
| | - Augusto Rojas-Martinez
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey, Nuevo Leon 64710, Mexico
| | - Juan Francisco Gonzalez-Guerrero
- Servicio de Oncologia, Centro Universitario Contra el Cancer, Hospital Universitario Dr. Jose Eleuterio Gonzalez, Universidad Autonoma de Nuevo Leon, Monterrey, Nuevo Leon 66460, Mexico
| | - Javier Valero-Gomez
- Centro de Cancer de Mama, Hospital San Jose, Tecnologico de Monterrey, Monterrey, Nuevo Leon 64710, Mexico
| | - Gabriela S Gomez-Macias
- Servicio de Patología, Hospital San Jose, Tecnologico de Monterrey, Hospital San Jose, Monterrey, Nuevo Leon 64710, Mexico
| | - Alvaro Barbosa-Quintana
- Servicio de Patología, Hospital San Jose, Tecnologico de Monterrey, Hospital San Jose, Monterrey, Nuevo Leon 64710, Mexico
| | - Oralia Barboza-Quintana
- Servicio de Anatomia Patologica y Citopatologia, Hospital Universitario Dr. Jose Eleuterio Gonzalez, Universidad Autonoma de Nuevo Leon, Monterrey, Nuevo Leon 66450, Mexico
| | - Raquel Garza-Guajardo
- Servicio de Anatomia Patologica y Citopatologia, Hospital Universitario Dr. Jose Eleuterio Gonzalez, Universidad Autonoma de Nuevo Leon, Monterrey, Nuevo Leon 66450, Mexico
| | - Rocio Ortiz-Lopez
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey, Nuevo Leon 64710, Mexico
| |
Collapse
|
77
|
CDK6 coordinates JAK2 V617F mutant MPN via NF-κB and apoptotic networks. Blood 2019; 133:1677-1690. [PMID: 30635286 DOI: 10.1182/blood-2018-08-872648] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/07/2019] [Indexed: 01/27/2023] Open
Abstract
Over 80% of patients with myeloproliferative neoplasms (MPNs) harbor the acquired somatic JAK2 V617F mutation. JAK inhibition is not curative and fails to induce a persistent response in most patients, illustrating the need for the development of novel therapeutic approaches. We describe a critical role for CDK6 in MPN evolution. The absence of Cdk6 ameliorates clinical symptoms and prolongs survival. The CDK6 protein interferes with 3 hallmarks of disease: besides regulating malignant stem cell quiescence, it promotes nuclear factor κB (NF-κB) signaling and contributes to cytokine production while inhibiting apoptosis. The effects are not mirrored by palbociclib, showing that the functions of CDK6 in MPN pathogenesis are largely kinase independent. Our findings thus provide a rationale for targeting CDK6 in MPN.
Collapse
|
78
|
Uras IZ, Maurer B, Nebenfuehr S, Zojer M, Valent P, Sexl V. Therapeutic Vulnerabilities in FLT3-Mutant AML Unmasked by Palbociclib. Int J Mol Sci 2018; 19:ijms19123987. [PMID: 30544932 PMCID: PMC6321303 DOI: 10.3390/ijms19123987] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 12/07/2018] [Indexed: 12/25/2022] Open
Abstract
While significant progress has been made in the treatment of acute myeloid leukemia (AML), not all patients can be cured. Mutated in about 1/3 of de novo AML, the FLT3 receptor tyrosine kinase is an attractive target for drug development, activating mutations of the FLT3 map to the juxtamembrane domain (internal tandem duplications, ITD) or the tyrosine kinase domain (TKD), most frequently at codon D835. While small molecule tyrosine kinase inhibitors (TKI) effectively target ITD mutant forms, those on the TKD are not responsive. Moreover, FLT3 inhibition fails to induce a persistent response in patients due to mutational resistance. More potent compounds with broader inhibitory effects on multiple FLT3 mutations are highly desirable. We describe a critical role of CDK6 in the survival of FLT3+ AML cells as palbociclib induced apoptosis not only in FLT3–ITD+ cells but also in FLT3–D835Y+ cells. Antineoplastic effects were also seen in primary patient-derived cells and in a xenograft model, where therapy effectively suppressed tumor formation in vivo at clinically relevant concentrations. In cells with FLT3–ITD or -TKD mutations, the CDK6 protein not only affects cell cycle progression but also transcriptionally regulates oncogenic kinases mediating intrinsic drug resistance, including AURORA and AKT—a feature not shared by its homolog CDK4. While AKT and AURORA kinase inhibitors have significant therapeutic potential in AML, single agent activity has not been proven overly effective. We describe synergistic combination effects when applying these drugs together with palbociclib which could be readily translated to patients with AML bearing FLT3–ITD or –TKD mutations. Targeting synergistically acting vulnerabilities, with CDK6 being the common denominator, may represent a promising strategy to improve AML patient responses and to reduce the incidence of selection of resistance-inducing mutations.
Collapse
Affiliation(s)
- Iris Z Uras
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria.
| | - Barbara Maurer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria.
| | - Sofie Nebenfuehr
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria.
| | - Markus Zojer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria.
| | - Peter Valent
- Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, 1090 Vienna, Austria.
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria.
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria.
| |
Collapse
|
79
|
Martinez-Soria N, McKenzie L, Draper J, Ptasinska A, Issa H, Potluri S, Blair HJ, Pickin A, Isa A, Chin PS, Tirtakusuma R, Coleman D, Nakjang S, Assi S, Forster V, Reza M, Law E, Berry P, Mueller D, Osborne C, Elder A, Bomken SN, Pal D, Allan JM, Veal GJ, Cockerill PN, Wichmann C, Vormoor J, Lacaud G, Bonifer C, Heidenreich O. The Oncogenic Transcription Factor RUNX1/ETO Corrupts Cell Cycle Regulation to Drive Leukemic Transformation. Cancer Cell 2018; 34:626-642.e8. [PMID: 30300583 PMCID: PMC6179967 DOI: 10.1016/j.ccell.2018.08.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 04/20/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022]
Abstract
Oncogenic transcription factors such as the leukemic fusion protein RUNX1/ETO, which drives t(8;21) acute myeloid leukemia (AML), constitute cancer-specific but highly challenging therapeutic targets. We used epigenomic profiling data for an RNAi screen to interrogate the transcriptional network maintaining t(8;21) AML. This strategy identified Cyclin D2 (CCND2) as a crucial transmitter of RUNX1/ETO-driven leukemic propagation. RUNX1/ETO cooperates with AP-1 to drive CCND2 expression. Knockdown or pharmacological inhibition of CCND2 by an approved drug significantly impairs leukemic expansion of patient-derived AML cells and engraftment in immunodeficient murine hosts. Our data demonstrate that RUNX1/ETO maintains leukemia by promoting cell cycle progression and identifies G1 CCND-CDK complexes as promising therapeutic targets for treatment of RUNX1/ETO-driven AML.
Collapse
Affiliation(s)
- Natalia Martinez-Soria
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Lynsey McKenzie
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Julia Draper
- Cancer Research UK Manchester Institute, Manchester M20 4GJ, UK
| | - Anetta Ptasinska
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Hasan Issa
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Sandeep Potluri
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Helen J Blair
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Anna Pickin
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Asmida Isa
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Paulynn Suyin Chin
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Ricky Tirtakusuma
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Daniel Coleman
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Sirintra Nakjang
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Salam Assi
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Victoria Forster
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Mojgan Reza
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Ed Law
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Philip Berry
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Dorothee Mueller
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Cameron Osborne
- Department of Medical & Molecular Genetics, King's College London, London SE1 9RT, UK
| | - Alex Elder
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Simon N Bomken
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Deepali Pal
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - James M Allan
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Gareth J Veal
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Peter N Cockerill
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Christian Wichmann
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, Ludwig-Maximilian University Hospital, Munich 80539, Germany
| | - Josef Vormoor
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK; Princess Maxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands
| | - Georges Lacaud
- Cancer Research UK Manchester Institute, Manchester M20 4GJ, UK
| | - Constanze Bonifer
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Olaf Heidenreich
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK; Princess Maxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands.
| |
Collapse
|
80
|
Nebenfuehr S, Bellutti F, Sexl V. Cdk6: At the interface of Rb and p53. Mol Cell Oncol 2018; 5:e1511206. [PMID: 30263948 PMCID: PMC6154857 DOI: 10.1080/23723556.2018.1511206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 08/07/2018] [Accepted: 08/09/2018] [Indexed: 02/08/2023]
Abstract
In cancer the activity of cyclin-dependent kinase 4-(CDK4) and cyclin-dependent kinase 6 (CDK6)-cyclin complexes are frequently altered with enhanced CDK6 expression found in hematopoietic malignancies. Our latest findings show a so far unknown role of Cdk6 during oncogene-induced stress and transformation. Therein Cdk6 antagonizes p53 responses and subsequently shapes the critical decision between survival and apoptosis in pre-leukemic cells.
Collapse
Affiliation(s)
- Sofie Nebenfuehr
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Florian Bellutti
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
81
|
Uras IZ, Bellutti F, Sexl V. p27 in FLT3-driven acute myeloid leukemia: many roads lead to ruin. Haematologica 2018; 102:1299-1301. [PMID: 28760806 DOI: 10.3324/haematol.2017.171819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Iris Z Uras
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Florian Bellutti
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
82
|
Bellutti F, Tigan AS, Nebenfuehr S, Dolezal M, Zojer M, Grausenburger R, Hartenberger S, Kollmann S, Doma E, Prchal-Murphy M, Uras IZ, Höllein A, Neuberg DS, Ebert BL, Ringler A, Mueller AC, Loizou JI, Hinds PW, Vogl C, Heller G, Kubicek S, Zuber J, Malumbres M, Farlik M, Villunger A, Kollmann K, Sexl V. CDK6 Antagonizes p53-Induced Responses during Tumorigenesis. Cancer Discov 2018; 8:884-897. [PMID: 29899063 DOI: 10.1158/2159-8290.cd-17-0912] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 04/05/2018] [Accepted: 05/08/2018] [Indexed: 02/07/2023]
Abstract
Tumor formation is a multistep process during which cells acquire genetic and epigenetic changes until they reach a fully transformed state. We show that CDK6 contributes to tumor formation by regulating transcriptional responses in a stage-specific manner. In early stages, the CDK6 kinase induces a complex transcriptional program to block p53 in hematopoietic cells. Cells lacking CDK6 kinase function are required to mutate TP53 (encoding p53) to achieve a fully transformed immortalized state. CDK6 binds to the promoters of genes including the p53 antagonists Prmt5, Ppm1d, and Mdm4 The findings are relevant to human patients: Tumors with low levels of CDK6 have mutations in TP53 significantly more often than expected.Significance: CDK6 acts at the interface of p53 and RB by driving cell-cycle progression and antagonizing stress responses. While sensitizing cells to p53-induced cell death, specific inhibition of CDK6 kinase activity may provoke the outgrowth of p53-mutant clones from premalignant cells. Cancer Discov; 8(7); 884-97. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 781.
Collapse
Affiliation(s)
- Florian Bellutti
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Anca-Sarmiza Tigan
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Sofie Nebenfuehr
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Marlies Dolezal
- Platform Bioinformatics and Biostatistics, University of Veterinary Medicine, Vienna, Austria
| | - Markus Zojer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Reinhard Grausenburger
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Svenja Hartenberger
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Sebastian Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Eszter Doma
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Michaela Prchal-Murphy
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Iris Z Uras
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | | | - Donna S Neuberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Benjamin L Ebert
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Anna Ringler
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Andre C Mueller
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Joanna I Loizou
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Philip W Hinds
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, and Tufts Cancer Center, Boston, Massachusetts
| | - Claus Vogl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | | | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Johannes Zuber
- Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | | | - Matthias Farlik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Karoline Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria.
| |
Collapse
|
83
|
Buetti-Dinh A, Friedman R. Computer simulations of the signalling network in FLT3 +-acute myeloid leukaemia - indications for an optimal dosage of inhibitors against FLT3 and CDK6. BMC Bioinformatics 2018; 19:155. [PMID: 29699481 PMCID: PMC5921566 DOI: 10.1186/s12859-018-2145-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 04/03/2018] [Indexed: 12/31/2022] Open
Abstract
Background Mutations in the FMS-like tyrosine kinase 3 (FLT3) are associated with uncontrolled cellular functions that contribute to the development of acute myeloid leukaemia (AML). We performed computer simulations of the FLT3-dependent signalling network in order to study the pathways that are involved in AML development and resistance to targeted therapies. Results Analysis of the simulations revealed the presence of alternative pathways through phosphoinositide 3 kinase (PI3K) and SH2-containing sequence proteins (SHC), that could overcome inhibition of FLT3. Inhibition of cyclin dependent kinase 6 (CDK6), a related molecular target, was also tested in the simulation but was not found to yield sufficient benefits alone. Conclusions The PI3K pathway provided a basis for resistance to treatments. Alternative signalling pathways could not, however, restore cancer growth signals (proliferation and loss of apoptosis) to the same levels as prior to treatment, which may explain why FLT3 resistance mutations are the most common resistance mechanism. Finally, sensitivity analysis suggested the existence of optimal doses of FLT3 and CDK6 inhibitors in terms of efficacy and toxicity. Electronic supplementary material The online version of this article (10.1186/s12859-018-2145-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Antoine Buetti-Dinh
- Department of Chemistry and Biomedical Sciences, Linnæus University, Norra vägen 49, Kalmar, SE-391 82, Sweden.,Linnæus University Centre for Biomaterials Chemistry, Linnæus University, Norra vägen 49, Kalmar, SE-391 82, Sweden.,Centre for Ecology and Evolution in Microbial Model Systems, Linnæus University, Landgången 3, Kalmar, SE-391 82, Sweden.,Institute of Computational Science, Faculty of Informatics, Università della Svizzera Italiana, Via Giuseppe Buffi 13, Lugano, CH-6900, Switzerland.,Swiss Institute of Bioinformatics, Quartier Sorge - Batiment Genopode, Lausanne, CH-1015, Switzerland
| | - Ran Friedman
- Department of Chemistry and Biomedical Sciences, Linnæus University, Norra vägen 49, Kalmar, SE-391 82, Sweden. .,Linnæus University Centre for Biomaterials Chemistry, Linnæus University, Norra vägen 49, Kalmar, SE-391 82, Sweden.
| |
Collapse
|
84
|
Ikeda S, Lim JS, Kurzrock R. Analysis of Tissue and Circulating Tumor DNA by Next-Generation Sequencing of Hepatocellular Carcinoma: Implications for Targeted Therapeutics. Mol Cancer Ther 2018; 17:1114-1122. [PMID: 29483209 DOI: 10.1158/1535-7163.mct-17-0604] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/21/2017] [Accepted: 02/12/2018] [Indexed: 12/15/2022]
Abstract
Hepatocellular carcinoma (HCC) has limited treatment options. Molecular analysis of its mutational landscape may enable the identification of novel therapies. However, biopsy is not routinely performed in HCC. The utility of analyzing cell-free circulating tumor DNA (ctDNA) by next-generation sequencing (NGS) is not established. We performed 32 ctDNA NGS analyses on 26 patients; 10 of these patients had tissue NGS (236 to 626 genes). ctDNA was evaluated using an assay that detects single nucleotide variants, amplifications, fusions, and specific insertion/deletion alterations in 54 to 70 genes. The ctDNA demonstrated that 23 of 26 patients (88.5%) had ≥1 characterized alteration, and all these individuals had ≥1 potentially actionable alteration. The most frequently mutated gene was TP53 (16 of 26 patients, 61.5%). There were 47 unique characterized molecular alterations among 18 total gene alterations [variants of unknown significance (VUS) excluded)]. ctDNA and tissue NGS frequently showed different profiles, perhaps due to length of time between tissue and blood samples [median = 370 days (range, 29 to 876 days)]. Serial ctDNA evaluation in an illustrative patient treated with capecitabine demonstrated emergence of a new TP53 alteration after progression. In conclusion, ctDNA profiling is feasible in advanced HCC, and serial assessment using ctDNA NGS can reveal genomic changes with time. NGS of ctDNA provides a minimally invasive alternative for identifying potentially actionable gene alterations and potential molecular targeted therapies. Dynamic changes in molecular portfolio associated with therapeutic pressure in difficult-to-biopsy patients can be observed. Mol Cancer Ther; 17(5); 1114-22. ©2018 AACR.
Collapse
Affiliation(s)
- Sadakatsu Ikeda
- Center for Personalized Cancer Therapy, Division of Hematology/Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla, California. .,Tokyo Medical and Dental University, Tokyo, Japan
| | - Jordan S Lim
- Center for Personalized Cancer Therapy, Division of Hematology/Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla, California.
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, Division of Hematology/Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla, California
| |
Collapse
|
85
|
Abstract
During the last decades, much has been learned about with cyclin-dependent kinases (CDK) playing a pivotal role in the cell cycle regulation. CDK4/6 is the key regulator of the G1-S transition. Palbociclib (PD 0332991, Ibrance®) is the first oral CDK4/6 inhibitor showing a substantially improved median progression-free survival (PFS) in advanced estrogen receptor (ER) positive and human epidermal growth factor receptor 2 (HER2) negative breast cancer. This PFS prolongation was seen both with letrozole as first-line therapy (24.8 vs. 14.5 months [PALOMA 2]) and with fulvestrant in endocrine pretreated patients (9.2 vs. 3.8 months [PALOMA-3]). The main toxicity is neutropenia due to cell cycle arrest which can be easily managed with dose interruption or dose reduction leading to a favorable safety profile with delayed deterioration of global quality of life (QoL). Palbociclib is approved by the Federal Drug Administration (FDA) and the European Medicines Agency (EMA) for ER-positive/HER2-negative advanced breast cancer. Despite the well-understood mode of action of palbociclib, predictive biomarkers are not yet defined. In conclusion, inhibition of CDK4/6 using palbociclib in combination with endocrine therapy is an efficient and well-tolerated treatment option in ER-positive/HER2-negative advanced breast cancer. Ongoing clinical trials are investigating the role of palbociclib in early breast cancer as well as in other types of cancer.
Collapse
Affiliation(s)
- Marcus Schmidt
- Department of Obstetrics and Gynecology, University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Martin Sebastian
- Department of Hematology/Oncology, Rheumatology, HIV, J.W. Goethe University, Frankfurt, Germany
| |
Collapse
|
86
|
He H, Xu J, Xie W, Guo QL, Jiang FL, Liu Y. Reduced state transition barrier of CDK6 from open to closed state induced by Thr177 phosphorylation and its implication in binding modes of inhibitors. Biochim Biophys Acta Gen Subj 2017; 1862:501-512. [PMID: 29108955 DOI: 10.1016/j.bbagen.2017.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/30/2017] [Accepted: 11/01/2017] [Indexed: 01/15/2023]
Abstract
BACKGROUND CDK6 is considered as a highly validated anticancer drug target due to its essential role in regulating cell cycle progression at G1 restriction point. Activation of CDK6 requires the phosphorylation of Thr177 on A-loop, but the structural insights of the activation mechanism remain unclear. METHODS Herein, all-atoms molecular dynamics (MD) simulations were used to study the effects of Thr177 phosphorylation on the dynamic structure of CDK6-Vcyclin complex. RESULTS MD results indicated that the free energy barrier of the transition from open to closed state decreased ~47.2% after Thr177 phosphorylation. Key steps along the state transition process were obtained from a cluster analysis. Binding preference of ten different inhibitors to open or closed state were also investigated through molecular docking along with MD simulations methods. CONCLUSIONS Our results indicated that Thr177 phosphorylation increased the flexibility around the ATP-binding pocket. The transition of the ATP-binding pocket between open and closed states should be considered for understanding the binding of CDK6 inhibitors. GENERAL SIGNIFICANCE This work could deepen the understanding of CDKs activation mechanism, and provide useful information for the discovery of new CDKs inhibitors with high affinity and specificity.
Collapse
Affiliation(s)
- Huan He
- State Key Laboratory of Virology & Key Laboratory of Analytical Chemistry for Biology and Medicine (MOE) & Key Laboratory of Biomedical Polymer Materials (MOE), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, PR China
| | - Juan Xu
- State Key Laboratory of Virology & Key Laboratory of Analytical Chemistry for Biology and Medicine (MOE) & Key Laboratory of Biomedical Polymer Materials (MOE), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, PR China
| | - Wen Xie
- Department of Clinical Laboratory, Zhongnan Hospital, Wuhan University, Wuhan 430071, PR China
| | - Qing-Lian Guo
- Department of Clinical Laboratory, Zhongnan Hospital, Wuhan University, Wuhan 430071, PR China
| | - Feng-Lei Jiang
- State Key Laboratory of Virology & Key Laboratory of Analytical Chemistry for Biology and Medicine (MOE) & Key Laboratory of Biomedical Polymer Materials (MOE), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, PR China.
| | - Yi Liu
- State Key Laboratory of Virology & Key Laboratory of Analytical Chemistry for Biology and Medicine (MOE) & Key Laboratory of Biomedical Polymer Materials (MOE), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, PR China; Key Laboratory of Coal Conversion and Carbon Materials of Hubei Province, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, PR China; College of Chemistry and Material Sciences, Guangxi Teachers Education University, Nanning 530001, PR China.
| |
Collapse
|
87
|
Peschel I, Podmirseg SR, Taschler M, Duyster J, Götze KS, Sill H, Nachbaur D, Jäkel H, Hengst L. FLT3 and FLT3-ITD phosphorylate and inactivate the cyclin-dependent kinase inhibitor p27 Kip1 in acute myeloid leukemia. Haematologica 2017; 102:1378-1389. [PMID: 28522571 PMCID: PMC5541872 DOI: 10.3324/haematol.2016.160101] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 05/08/2017] [Indexed: 02/01/2023] Open
Abstract
P27 Kip1 (p27) can prevent cell proliferation by inactivating cyclin-dependent kinases. This function is impaired upon phosphorylation of p27 at tyrosine residue 88. We observed that FLT3 and FLT3-ITD can directly bind and selectively phosphorylate p27 on this residue. Inhibition of FLT3-ITD in cell lines strongly reduced p27 tyrosine 88 phosphorylation and resulted in increased p27 levels and cell cycle arrest. Subsequent analysis revealed the presence of tyrosine 88 phosphorylated p27 in primary patient samples. Inhibition of FLT3 kinase activity with AC220 significantly reduced p27 tyrosine 88 phosphorylation in cells isolated from FLT3 wild type expressing acute myeloid leukemia (AML) patients. In FLT3-ITD positive AML patients, p27 tyrosine 88 phosphorylation was reduced in 5 out of 9 subjects, but, surprisingly, was increased in 4 patients. This indicated that other tyrosine kinases such as Src family kinases might contribute to p27 tyrosine 88 phosphorylation in FLT3-ITD positive AML cells. In fact, incubation with the Src family kinase inhibitor dasatinib could decrease p27 tyrosine 88 phosphorylation in these patient samples, indicating that p27 phosphorylated on tyrosine 88 may be a therapeutic marker for the treatment of AML patients with tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Ines Peschel
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Austria
| | - Silvio R Podmirseg
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Austria
| | - Martin Taschler
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Austria
| | - Justus Duyster
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, Germany
| | - Katharina S Götze
- Department of Internal Medicine III, Klinikum Rechts der Isar, Technical University of Munich, Germany
| | - Heinz Sill
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, Austria
| | - David Nachbaur
- Department of Internal Medicine V, Medical University of Innsbruck, Austria
| | - Heidelinde Jäkel
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Austria
| | - Ludger Hengst
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Austria
| |
Collapse
|
88
|
Abstract
INTRODUCTION AML therapy remains very challenging despite our increased understanding of its molecular heterogeneity. Outcomes with chemotherapy and targeted therapy remain poor. Targeting cell cycle regulators might complement chemotherapy and targeted therapy and help in improving outcomes. Areas covered: Here we cover the pre-clinical and clinical data for both for cyclin dependent kinase (CDK) and cell-cycle checkpoint inhibitors. While CDK inhibition can inhibit proliferation, checkpoint inhibitors can facilitate cell cycle progression in presence of DNA damage and can induce mitotic catastrophe. Expert opinion: Though the preclinical data for cell cycle inhibitors in AML is compelling, the clinical translation so far has proven to be challenging. This is a reflection of the complexity of both, AML and cell cycle regulators. However, early introduction of cell-cycle active agents in combination with chemotherapy or targeted agents, identifying right sequence of use and identifying right biomarkers might pave the way into successful clinical translation.
Collapse
Affiliation(s)
- Abdallah Abou Zahr
- a Department of Leukemia , University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Gautam Borthakur
- a Department of Leukemia , University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
89
|
Ghiaur G, Levis M. Mechanisms of Resistance to FLT3 Inhibitors and the Role of the Bone Marrow Microenvironment. Hematol Oncol Clin North Am 2017; 31:681-692. [PMID: 28673395 DOI: 10.1016/j.hoc.2017.04.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The presence of FLT3 mutations in acute myeloid leukemia (AML) carries a particularly poor prognosis, making the development of FLT3 inhibitors an imperative goal. The last decade has seen an abundance of clinical trials using these drugs alone or in combination with chemotherapy. This culminated with the recent approval by the US Food and Drug Administration of Midostaurin for the treatment of FLT3-mutated AML. Initial success has been followed by the emergence of clinical resistance. Although novel FLT3 inhibitors are being developed, studies into mechanisms of resistance raise hope of new strategies to prevent emergence of resistance and eliminate minimal residual disease.
Collapse
Affiliation(s)
- Gabriel Ghiaur
- Adult Leukemia Program, Division of Hematological Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, 1650 Orleans Street CRB I, Room 243, Baltimore, MD 21287, USA.
| | - Mark Levis
- Adult Leukemia Program, Division of Hematological Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, 1650 Orleans Street CRB I, Room 2M44, Baltimore, MD 21287, USA
| |
Collapse
|
90
|
Karathedath S, Rajamani BM, Musheer Aalam SM, Abraham A, Varatharajan S, Krishnamurthy P, Mathews V, Velayudhan SR, Balasubramanian P. Role of NF-E2 related factor 2 (Nrf2) on chemotherapy resistance in acute myeloid leukemia (AML) and the effect of pharmacological inhibition of Nrf2. PLoS One 2017; 12:e0177227. [PMID: 28505160 PMCID: PMC5432104 DOI: 10.1371/journal.pone.0177227] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 04/24/2017] [Indexed: 12/30/2022] Open
Abstract
Cytarabine (Ara-C) and Daunorubicin (Dnr) forms the backbone of acute myeloid leukemia (AML) therapy. Drug resistance and toxic side effects pose a major threat to treatment success and hence alternate less toxic therapies are warranted. NF-E2 related factor-2 (Nrf2), a master regulator of antioxidant response is implicated in chemoresistance in solid tumors. However, little is known about the role of Nrf2 in AML chemoresistance and the effect of pharmacological inhibitor brusatol in modulating this resistance. Primary AML samples with high ex-vivo IC50 to Ara-C, ATO, Dnr had significantly high NRF2 RNA expression. Gene-specific knockdown of NRF2 improved sensitivity to these drugs in resistant AML cell lines by decreasing the expression of downstream antioxidant targets of Nrf2 by compromising the cell’s ability to scavenge the ROS. Treatment with brusatol, a pharmacological inhibitor of Nrf2, improved sensitivity to Ara-C, ATO, and Dnr and reduced colony formation capacity. AML cell lines stably overexpressing NRF2 showed increased resistance to ATO, Dnr and Ara-C and increased expression of downstream targets. This study demonstrates that Nrf2 could be an ideal druggable target in AML, more so to the drugs that function through ROS, suggesting the possibility of using Nrf2 inhibitors in combination with chemotherapeutic agents to modulate drug resistance in AML.
Collapse
Affiliation(s)
| | | | | | - Ajay Abraham
- Department of Haematology, Christian Medical College, Vellore, India
| | | | - Partha Krishnamurthy
- Department of Pharmacology, Toxicology and Therapeutics, Kansas University Medical Centre, Kansas City, Kansas, United States of America
| | - Vikram Mathews
- Department of Haematology, Christian Medical College, Vellore, India
| | - Shaji Ramachandran Velayudhan
- Department of Haematology, Christian Medical College, Vellore, India
- Centre for Stem Cell Research, Christian Medical College, Vellore, India
| | | |
Collapse
|
91
|
|
92
|
Di Giovanni C, Novellino E, Chilin A, Lavecchia A, Marzaro G. Investigational drugs targeting cyclin-dependent kinases for the treatment of cancer: an update on recent findings (2013-2016). Expert Opin Investig Drugs 2017; 25:1215-30. [PMID: 27606939 DOI: 10.1080/13543784.2016.1234603] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Cell cycle and gene transcription are under the control of cyclin-dependent kinases (CDKs), whose activity depends on the binding with cyclins. Deregulated CDK activities have been reported in a majority of human cancers, representing potential therapeutic targets. AREAS COVERED This review provides preclinical and clinical (phase I/II) updates of promising therapeutic compounds targeting CDKs published between 2013 and 2016 EXPERT OPINION: First generation pan-CDK inhibitors showed marked toxicity in clinical trials and most compounds were discontinued. Despite their failure was ascribed also to inadequate patient selection rules, novel pan-CDK inhibitors have entered clinical trials with still poorly defined selection strategies. The most interesting results have been obtained with dual CDK4/6 inhibitors and through a more accurate evaluation of predictive biomarkers, suggesting the usefulness of CDK inhibitors for personalized treatment. The increased knowledge on the roles of CDKs in cell cycle and gene transcription suggests to review also the anticancer potential of first generation CDK inhibitors by defining more appropriate rules for patients engagement. Recent findings has highlighted CDK8 as a novel target for cancer treatment. Indeed some biomarkers for CDK8 inhibition sensitivity have already been proposed. CDK8 inhibition is also supposed to prevent cancer metastasis.
Collapse
Affiliation(s)
- Carmen Di Giovanni
- a Department of Pharmacy , University of Naples Federico II , Naples , Italy
| | - Ettore Novellino
- a Department of Pharmacy , University of Naples Federico II , Naples , Italy
| | - Adriana Chilin
- b Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Padova , Italy
| | - Antonio Lavecchia
- a Department of Pharmacy , University of Naples Federico II , Naples , Italy
| | - Giovanni Marzaro
- b Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Padova , Italy
| |
Collapse
|
93
|
Uras IZ, Scheicher RM, Kollmann K, Glösmann M, Prchal-Murphy M, Tigan AS, Fux DA, Altamura S, Neves J, Muckenthaler MU, Bennett KL, Kubicek S, Hinds PW, von Lindern M, Sexl V. Cdk6 contributes to cytoskeletal stability in erythroid cells. Haematologica 2017; 102:995-1005. [PMID: 28255017 PMCID: PMC5451331 DOI: 10.3324/haematol.2016.159947] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/22/2017] [Indexed: 12/03/2022] Open
Abstract
Mice lacking Cdk6 kinase activity suffer from mild anemia accompanied by elevated numbers of Ter119+ cells in the bone marrow. The animals show hardly any alterations in erythroid development, indicating that Cdk6 is not required for proliferation and maturation of erythroid cells. There is also no difference in stress erythropoiesis following hemolysis in vivo. However, Cdk6−/− erythrocytes have a shortened lifespan and are more sensitive to mechanical stress in vitro, suggesting differences in cytoskeletal architecture. Erythroblasts contain both Cdk4 and Cdk6, while mature erythrocytes apparently lack Cdk4 and their Cdk6 is partly associated with the cytoskeleton. We used mass spectrometry to show that Cdk6 interacts with a number of proteins involved in cytoskeleton organization. Cdk6−/− erythroblasts show impaired F-actin formation and lower levels of gelsolin, which interacts with Cdk6. We also found that Cdk6 regulates the transcription of a panel of genes involved in actin (de-)polymerization. Cdk6-deficient cells are sensitive to drugs that interfere with the cytoskeleton, suggesting that our findings are relevant to the treatment of patients with anemia – and may be relevant to cancer patients treated with the new generation of CDK6 inhibitors.
Collapse
Affiliation(s)
- Iris Z Uras
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Ruth M Scheicher
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Karoline Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | | | - Michaela Prchal-Murphy
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Anca S Tigan
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Daniela A Fux
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Sandro Altamura
- Department of Pediatric Hematology, Oncology, and Immunology, University of Heidelberg, Germany.,Molecular Medicine Partnership Unit, University of Heidelberg, Germany
| | - Joana Neves
- Department of Pediatric Hematology, Oncology, and Immunology, University of Heidelberg, Germany.,Molecular Medicine Partnership Unit, University of Heidelberg, Germany
| | - Martina U Muckenthaler
- Department of Pediatric Hematology, Oncology, and Immunology, University of Heidelberg, Germany.,Molecular Medicine Partnership Unit, University of Heidelberg, Germany
| | - Keiryn L Bennett
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Philip W Hinds
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, and Tufts Cancer Center, Boston, MA, USA
| | | | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
94
|
Tadesse S, Yu M, Mekonnen LB, Lam F, Islam S, Tomusange K, Rahaman MH, Noll B, Basnet SKC, Teo T, Albrecht H, Milne R, Wang S. Highly Potent, Selective, and Orally Bioavailable 4-Thiazol-N-(pyridin-2-yl)pyrimidin-2-amine Cyclin-Dependent Kinases 4 and 6 Inhibitors as Anticancer Drug Candidates: Design, Synthesis, and Evaluation. J Med Chem 2017; 60:1892-1915. [PMID: 28156111 DOI: 10.1021/acs.jmedchem.6b01670] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cyclin D dependent kinases (CDK4 and CDK6) regulate entry into S phase of the cell cycle and are validated targets for anticancer drug discovery. Herein we detail the discovery of a novel series of 4-thiazol-N-(pyridin-2-yl)pyrimidin-2-amine derivatives as highly potent and selective inhibitors of CDK4 and CDK6. Medicinal chemistry optimization resulted in 83, an orally bioavailable inhibitor molecule with remarkable selectivity. Repeated oral administration of 83 caused marked inhibition of tumor growth in MV4-11 acute myeloid leukemia mouse xenografts without having a negative effect on body weight and showing any sign of clinical toxicity. The data merit 83 as a clinical development candidate.
Collapse
Affiliation(s)
- Solomon Tadesse
- Center for Drug Discovery and Development, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, and Center for Cancer Biology, University of South Australia , Adelaide, South Australia 5001, Australia
| | - Mingfeng Yu
- Center for Drug Discovery and Development, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, and Center for Cancer Biology, University of South Australia , Adelaide, South Australia 5001, Australia
| | - Laychiluh B Mekonnen
- Center for Drug Discovery and Development, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, and Center for Cancer Biology, University of South Australia , Adelaide, South Australia 5001, Australia
| | - Frankie Lam
- Center for Drug Discovery and Development, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, and Center for Cancer Biology, University of South Australia , Adelaide, South Australia 5001, Australia
| | - Saiful Islam
- Center for Drug Discovery and Development, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, and Center for Cancer Biology, University of South Australia , Adelaide, South Australia 5001, Australia
| | - Khamis Tomusange
- Center for Drug Discovery and Development, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, and Center for Cancer Biology, University of South Australia , Adelaide, South Australia 5001, Australia
| | - Muhammed H Rahaman
- Center for Drug Discovery and Development, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, and Center for Cancer Biology, University of South Australia , Adelaide, South Australia 5001, Australia
| | - Benjamin Noll
- Center for Drug Discovery and Development, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, and Center for Cancer Biology, University of South Australia , Adelaide, South Australia 5001, Australia
| | - Sunita K C Basnet
- Center for Drug Discovery and Development, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, and Center for Cancer Biology, University of South Australia , Adelaide, South Australia 5001, Australia
| | - Theodosia Teo
- Center for Drug Discovery and Development, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, and Center for Cancer Biology, University of South Australia , Adelaide, South Australia 5001, Australia
| | - Hugo Albrecht
- Center for Drug Discovery and Development, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, and Center for Cancer Biology, University of South Australia , Adelaide, South Australia 5001, Australia
| | - Robert Milne
- Center for Drug Discovery and Development, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, and Center for Cancer Biology, University of South Australia , Adelaide, South Australia 5001, Australia
| | - Shudong Wang
- Center for Drug Discovery and Development, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, and Center for Cancer Biology, University of South Australia , Adelaide, South Australia 5001, Australia
| |
Collapse
|