51
|
Gugliandolo A, Rajan TS, Scionti D, Diomede F, Bramanti P, Mazzon E, Trubiani O. Reprogramming of Oncogene Expression in Gingival Mesenchymal Stem Cells Following Long-Term Culture In Vitro. Cell Reprogram 2017; 19:159-170. [DOI: 10.1089/cell.2016.0056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
| | | | | | - Francesca Diomede
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University “G. d'Annunzio”, Chieti-Pescara, Chieti, Italy
| | | | | | - Oriana Trubiani
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University “G. d'Annunzio”, Chieti-Pescara, Chieti, Italy
| |
Collapse
|
52
|
Lee JW, Park SH, Kang HJ, Park KD, Shin HY, Ahn HS. ALK Protein Expression Is Related to Neuroblastoma Aggressiveness But Is Not Independent Prognostic Factor. Cancer Res Treat 2017; 50:495-505. [PMID: 28546523 PMCID: PMC5912141 DOI: 10.4143/crt.2016.577] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 05/16/2017] [Indexed: 12/11/2022] Open
Abstract
PURPOSE In this study, anaplastic lymphoma kinase (ALK) mutation and amplification, ALK protein expression, loss of the nuclear alpha thalassemia/mental retardation syndrome X-linked (ATRX) protein, and telomerase reverse transcriptase (TERT) protein expressionwere studied to investigate potential correlations between these molecular characteristics and clinical features or outcomes in neuroblastoma. MATERIALS AND METHODS Seventy-two patients were enrolled in this study. Polymerase chain reaction amplification and direct sequencing were used for mutation analysis. ALK and MYCN amplifications were detected by fluorescence in situ hybridization. Protein expressionwas evaluated by immunohistochemical (IHC) staining. RESULTS ALK mutation was found in only two patients (4.1%); ALK amplification was not detected. ALK positivity, loss of nuclear ATRX protein, TERT positivity by IHC were detected in 40 (55.6%), nine (13.0%), and 42 (59.2%) patients, respectively. The incidence of ALK expression increased in accordance with increasing tumor stage (p=0.001) and risk group (p < 0.001). The relapse rate was significantly higher in ALK+ patients compared to that of other patients (47.5% vs. 11.3%, p=0.007). However, there was no significant difference in relapse rate when the survival analysis was confined to the high-risk patients. CONCLUSION Although ALK mutation was rare and no amplification was observed, ALK protein expression was found in a significant number of patients and was correlated with advanced stage and high-risk neuroblastoma. ALK protein expression could be considered as a marker related to the aggressive neuroblastoma, but it was not the independent prognostic factor for the outcome.
Collapse
Affiliation(s)
- Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung Hye Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Hyoung Jin Kang
- Department of Pediatrics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung Duk Park
- Department of Pediatrics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hee Young Shin
- Department of Pediatrics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyo Seop Ahn
- Department of Pediatrics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
53
|
Hoang VT, Yan TJ, Cavanaugh JE, Flaherty PT, Beckman BS, Burow ME. Oncogenic signaling of MEK5-ERK5. Cancer Lett 2017; 392:51-59. [PMID: 28153789 PMCID: PMC5901897 DOI: 10.1016/j.canlet.2017.01.034] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 01/17/2017] [Accepted: 01/23/2017] [Indexed: 12/17/2022]
Abstract
Mitogen-activated protein kinases (MAPKs) regulate diverse cellular processes including proliferation, cell survival, differentiation, and apoptosis. While conventional MAPK constituents have well-defined roles in oncogenesis, the MEK5 pathway has only recently emerged in cancer research. In this review, we consider the MEK5 signaling cascade, focusing specifically on its involvement in drug resistance and regulation of aggressive cancer phenotypes. Moreover, we explore the role of MEK5/ERK5 in tumorigenesis and metastatic progression, discussing the discrepancies in preclinical studies and assessing its viability as a therapeutic target for anti-cancer agents.
Collapse
Affiliation(s)
- Van T Hoang
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA
| | - Thomas J Yan
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA
| | - Jane E Cavanaugh
- Department of Pharmacological Sciences, Division of Medicinal Chemistry, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - Patrick T Flaherty
- Department of Pharmacological Sciences, Division of Medicinal Chemistry, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | | | - Matthew E Burow
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA; Department of Pharmacology, Tulane University, New Orleans, LA, USA; Tulane Cancer Center, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
54
|
Speleman F, Park JR, Henderson TO. Neuroblastoma: A Tough Nut to Crack. Am Soc Clin Oncol Educ Book 2017; 35:e548-57. [PMID: 27249766 DOI: 10.1200/edbk_159169] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neuroblastoma, an embryonal tumor arising from neural crest-derived progenitor cells, is the most common solid tumor in childhood, with more than 700 cases diagnosed per year in the United States. In the past several decades, significant advances have been made in the treatment of neuroblastoma. Treatment advances reflect improved understanding of the biology of neuroblastoma. Although amplification of MYCN was discovered in the early 1980s, our understanding of neuroblastoma oncogenesis has advanced in the last decade as a result of high-throughput genomic analysis, exome and whole-genome sequencing, genome-wide association studies, and synthetic lethal drug screens. Our refined understanding of neuroblastoma biology and genetics is reflected in improved prognostic stratification and appropriate tailoring of therapy in recent clinical trials. Moreover, for high-risk neuroblastoma, a disease that was uniformly fatal 3 decades ago, recent clinical trials incorporating autologous hematopoietic transplant and immunotherapy utilizing anti-GD2 antibody plus cytokines have shown improved event-free and overall survival. These advances have resulted in a growing population of long-term survivors of neuroblastoma. Examination of the late effects and second malignant neoplasms (SMNs) in both older generations of survivors and more recently treated survivors will inform both design of future trials and surveillance guidelines for long-term follow-up. As a consequence of advances in understanding of the biology of neuroblastoma, successful clinical trials, and refined understanding of the late effects and SMNs of survivors, the promise of precision medicine is becoming a reality for patients with neuroblastoma.
Collapse
Affiliation(s)
- Frank Speleman
- From the Center for Medical Genetics Ghent, Cancer Research Institute Ghent, Ghent, Belgium; Seattle Children's Hospital, Seattle, WA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA; University of Chicago Comer Children's Hospital, Chicago, IL
| | - Julie R Park
- From the Center for Medical Genetics Ghent, Cancer Research Institute Ghent, Ghent, Belgium; Seattle Children's Hospital, Seattle, WA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA; University of Chicago Comer Children's Hospital, Chicago, IL
| | - Tara O Henderson
- From the Center for Medical Genetics Ghent, Cancer Research Institute Ghent, Ghent, Belgium; Seattle Children's Hospital, Seattle, WA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA; University of Chicago Comer Children's Hospital, Chicago, IL
| |
Collapse
|
55
|
Liu F, Zhang H, Song H. Upregulation of MEK5 by Stat3 promotes breast cancer cell invasion and metastasis. Oncol Rep 2017; 37:83-90. [PMID: 27878304 DOI: 10.3892/or.2016.5256] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 10/03/2016] [Indexed: 02/05/2023] Open
Abstract
Mitogen extracellular-signal-regulated kinase kinase 5 (MEK5) plays an important role in promoting cell proliferation and tumorigenesis. The aberrant expression of MEK5 has been reported in various malignant diseases including cancers of breast, prostate, lung, colorectal and brain. However, the function and regulation of MEK5 signaling pathway are ambiguous and remain elusive with respect to its oncogenic roles in various cancers, especially in the regulation of the initiation and progression of cancer invasion and metastasis. Ectopic expression of MEK5 or knockdown of MEK5 by shRNA with in vitro cell based models demonstrated the role of MEK5 in regulation of epithelial mesenchymal transition (EMT) and breast cancer invasion and metastasis. Here, we show that MEK5 upregulated by Stat3 promotes breast cancer cell invasion through EMT. Further study demonstrated that Stat3 could bind to promoter region of MEK5 and enhanced MEK5 transcription and expression. In addition, the phosphorylation of MEK5 significantly increased in breast cancer cells corresponding to metastatic capability of breast cancer cells. The depletion of MEK5 by shRNA significantly decreased breast cancer invasion. Ectopic expression of MEK5 could confer non-invasive breast cancer cells to become invasion capable cells. Moreover, the phosphorylation of Erk5, a MEK5-regulated downstream kinase, was also upregulated consistent with the increased level of active MEK5. Our studies provide insights into a molecular mechanism by which MEK5 transcriptionally upregulated by Stat3 augments breast cancer cell EMT, which subsequently enhances cancer cell invasion and metastasis. This finding may suggest that Stat3 and MEK5/Erk5 pathways could be an effective therapeutic target for inhibition of breast cancer invasion and metastasis.
Collapse
Affiliation(s)
- Fang Liu
- Xi'an Jiaotong University Suzhou Academy, Suzhou, Jiangsu 215123, P.R. China
| | - Hao Zhang
- Shantou University Medical College Cancer Research Center, Shantou, Guangdong 515041, P.R. China
| | - Hui Song
- Xi'an Jiaotong University Suzhou Academy, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
56
|
Nelson KN, Peiris MN, Meyer AN, Siari A, Donoghue DJ. Receptor Tyrosine Kinases: Translocation Partners in Hematopoietic Disorders. Trends Mol Med 2016; 23:59-79. [PMID: 27988109 DOI: 10.1016/j.molmed.2016.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/11/2016] [Accepted: 11/13/2016] [Indexed: 02/07/2023]
Abstract
Receptor tyrosine kinases (RTKs) activate various signaling pathways and regulate cellular proliferation, survival, migration, and angiogenesis. Malignant neoplasms often circumvent or subjugate these pathways by promoting RTK overactivation through mutation or chromosomal translocation. RTK translocations create a fusion protein containing a dimerizing partner fused to an RTK kinase domain, resulting in constitutive kinase domain activation, altered RTK cellular localization, upregulation of downstream signaling, and novel pathway activation. While RTK translocations in hematological malignancies are relatively rare, clinical evidence suggests that patients with these genetic abnormalities benefit from RTK-targeted inhibitors. Here, we present a timely review of an exciting field by examining RTK chromosomal translocations in hematological cancers, such as Anaplastic Lymphoma Kinase (ALK), Fibroblast Growth Factor Receptor (FGFR), Platelet-Derived Growth Factor Receptor (PDGFR), REarranged during Transfection (RET), Colony Stimulating Factor 1 Receptor (CSF1R), and Neurotrophic Tyrosine Kinase Receptor Type 3 (NTRK3) fusions, and discuss current therapeutic options.
Collapse
Affiliation(s)
- Katelyn N Nelson
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Malalage N Peiris
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - April N Meyer
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Asma Siari
- Université Joseph Fourier Grenoble, Grenoble, France
| | - Daniel J Donoghue
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA; Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
57
|
Duffy DJ, Krstic A, Halasz M, Schwarzl T, Fey D, Iljin K, Mehta JP, Killick K, Whilde J, Turriziani B, Haapa-Paananen S, Fey V, Fischer M, Westermann F, Henrich KO, Bannert S, Higgins DG, Kolch W. Integrative omics reveals MYCN as a global suppressor of cellular signalling and enables network-based therapeutic target discovery in neuroblastoma. Oncotarget 2016; 6:43182-201. [PMID: 26673823 PMCID: PMC4791225 DOI: 10.18632/oncotarget.6568] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 11/23/2015] [Indexed: 12/12/2022] Open
Abstract
Despite intensive study, many mysteries remain about the MYCN oncogene's functions. Here we focus on MYCN's role in neuroblastoma, the most common extracranial childhood cancer. MYCN gene amplification occurs in 20% of cases, but other recurrent somatic mutations are rare. This scarcity of tractable targets has hampered efforts to develop new therapeutic options. We employed a multi-level omics approach to examine MYCN functioning and identify novel therapeutic targets for this largely un-druggable oncogene. We used systems medicine based computational network reconstruction and analysis to integrate a range of omic techniques: sequencing-based transcriptomics, genome-wide chromatin immunoprecipitation, siRNA screening and interaction proteomics, revealing that MYCN controls highly connected networks, with MYCN primarily supressing the activity of network components. MYCN's oncogenic functions are likely independent of its classical heterodimerisation partner, MAX. In particular, MYCN controls its own protein interaction network by transcriptionally regulating its binding partners. Our network-based approach identified vulnerable therapeutically targetable nodes that function as critical regulators or effectors of MYCN in neuroblastoma. These were validated by siRNA knockdown screens, functional studies and patient data. We identified β-estradiol and MAPK/ERK as having functional cross-talk with MYCN and being novel targetable vulnerabilities of MYCN-amplified neuroblastoma. These results reveal surprising differences between the functioning of endogenous, overexpressed and amplified MYCN, and rationalise how different MYCN dosages can orchestrate cell fate decisions and cancerous outcomes. Importantly, this work describes a systems-level approach to systematically uncovering network based vulnerabilities and therapeutic targets for multifactorial diseases by integrating disparate omic data types.
Collapse
Affiliation(s)
- David J Duffy
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland.,The Whitney Laboratory for Marine Bioscience, University of Florida, St. Augustine, Florida, USA
| | - Aleksandar Krstic
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Melinda Halasz
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Thomas Schwarzl
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland.,European Molecular Biology Laboratory (EMBL), Meyerhofstraße, Heidelberg, Germany
| | - Dirk Fey
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | | | - Jai Prakash Mehta
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Kate Killick
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Jenny Whilde
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | | | | | - Vidal Fey
- VTT Technical Research Centre of Finland, Espoo, Finland
| | - Matthias Fischer
- Department of Paediatric Haematology and Oncology and Center for Molecular Medicine Cologne (CMMC), University Hospital Cologne, Cologne, Germany
| | - Frank Westermann
- Division of NB Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kai-Oliver Henrich
- Division of NB Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Steffen Bannert
- Division of NB Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Desmond G Higgins
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland.,Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Walter Kolch
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland.,Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
58
|
Gomez N, Erazo T, Lizcano JM. ERK5 and Cell Proliferation: Nuclear Localization Is What Matters. Front Cell Dev Biol 2016; 4:105. [PMID: 27713878 PMCID: PMC5031611 DOI: 10.3389/fcell.2016.00105] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 09/06/2016] [Indexed: 12/31/2022] Open
Abstract
ERK5, the last MAP kinase family member discovered, is activated by the upstream kinase MEK5 in response to growth factors and stress stimulation. MEK5-ERK5 pathway has been associated to different cellular processes, playing a crucial role in cell proliferation in normal and cancer cells by mechanisms that are both dependent and independent of its kinase activity. Thus, nuclear ERK5 activates transcription factors by either direct phosphorylation or acting as co-activator thanks to a unique transcriptional activation TAD domain located at its C-terminal tail. Consequently, ERK5 has been proposed as an interesting target to tackle different cancers, and either inhibitors of ERK5 activity or silencing the protein have shown antiproliferative activity in cancer cells and to block tumor growth in animal models. Here, we review the different mechanisms involved in ERK5 nuclear translocation and their consequences. Inactive ERK5 resides in the cytosol, forming a complex with Hsp90-Cdc37 superchaperone. In a canonical mechanism, MEK5-dependent activation results in ERK5 C-terminal autophosphorylation, Hsp90 dissociation, and nuclear translocation. This mechanism integrates signals such as growth factors and stresses that activate the MEK5-ERK5 pathway. Importantly, two other mechanisms, MEK5-independent, have been recently described. These mechanisms allow nuclear shuttling of kinase-inactive forms of ERK5. Although lacking kinase activity, these forms activate transcription by interacting with transcription factors through the TAD domain. Both mechanisms also require Hsp90 dissociation previous to nuclear translocation. One mechanism involves phosphorylation of the C-terminal tail of ERK5 by kinases that are activated during mitosis, such as Cyclin-dependent kinase-1. The second mechanism involves overexpression of chaperone Cdc37, an oncogene that is overexpressed in cancers such as prostate adenocarcinoma, where it collaborates with ERK5 to promote cell proliferation. Although some ERK5 kinase inhibitors have shown antiproliferative activity it is likely that those tumors expressing kinase-inactive nuclear ERK5 will not respond to these inhibitors.
Collapse
Affiliation(s)
| | | | - Jose M. Lizcano
- Protein Kinases and Signal Transduction Laboratory, Institut de Neurociencies and Departament de Bioquimica i Biologia Molecular, Facultat de Medicina, Universitat Autonoma de BarcelonaBarcelona, Spain
| |
Collapse
|
59
|
Satoh S, Takatori A, Ogura A, Kohashi K, Souzaki R, Kinoshita Y, Taguchi T, Hossain MS, Ohira M, Nakamura Y, Nakagawara A. Neuronal leucine-rich repeat 1 negatively regulates anaplastic lymphoma kinase in neuroblastoma. Sci Rep 2016; 6:32682. [PMID: 27604320 PMCID: PMC5015029 DOI: 10.1038/srep32682] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 08/03/2016] [Indexed: 12/12/2022] Open
Abstract
In neuroblastoma (NB), one of the most common paediatric solid tumours, activation of anaplastic lymphoma kinase (ALK) is often associated with poor outcomes. Although genetic studies have identified copy number alteration and nonsynonymous mutations of ALK, the regulatory mechanism of ALK signalling at protein levels is largely elusive. Neuronal leucine-rich repeat 1 (NLRR1) is a type 1 transmembrane protein that is highly expressed in unfavourable NB and potentially influences receptor tyrosine kinase signalling. Here, we showed that NLRR1 and ALK exhibited a mutually exclusive expression pattern in primary NB tissues by immunohistochemistry. Moreover, dorsal root ganglia of Nlrr1+/+ and Nlrr1−/− mice displayed the opposite expression patterns of Nlrr1 and Alk. Of interest, NLRR1 physically interacted with ALK in vitro through its extracellular region. Notably, the NLRR1 ectodomain impaired ALK phosphorylation and proliferation of ALK-mutated NB cells. A newly identified cleavage of the NLRR1 ectodomain also supported NLRR1-mediated ALK signal regulation in trans. Thus, we conclude that NLRR1 appears to be an extracellular negative regulator of ALK signalling in NB and neuronal development. Our findings may be beneficial to comprehend NB heterogeneity and to develop a novel therapy against unfavourable NB.
Collapse
Affiliation(s)
- Shunpei Satoh
- Children's Cancer Research Center, Chiba Cancer Center Research Institute, Chiba 260-8717, Japan.,Department of Molecular Biology and Oncology, Graduate School of Medical and Pharmaceutical Sciences, Chiba University, Chiba 260-8670, Japan
| | - Atsushi Takatori
- Children's Cancer Research Center, Chiba Cancer Center Research Institute, Chiba 260-8717, Japan
| | - Atsushi Ogura
- Children's Cancer Research Center, Chiba Cancer Center Research Institute, Chiba 260-8717, Japan.,Department of Molecular Biology and Oncology, Graduate School of Medical and Pharmaceutical Sciences, Chiba University, Chiba 260-8670, Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology, Graduate School Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan, Chiba 260-8670, Japan
| | - Ryota Souzaki
- Department of Pediatric Surgery, Graduate School Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshiaki Kinoshita
- Department of Pediatric Surgery, Graduate School Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tomoaki Taguchi
- Department of Pediatric Surgery, Graduate School Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Md Shamim Hossain
- Children's Cancer Research Center, Chiba Cancer Center Research Institute, Chiba 260-8717, Japan
| | - Miki Ohira
- Division of Cancer Genomics, Chiba Cancer Center Research Institute, Chiba 260-8717, Japan
| | - Yohko Nakamura
- Division of Biochemistry &Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chiba 260-8717, Japan
| | - Akira Nakagawara
- Children's Cancer Research Center, Chiba Cancer Center Research Institute, Chiba 260-8717, Japan.,Department of Molecular Biology and Oncology, Graduate School of Medical and Pharmaceutical Sciences, Chiba University, Chiba 260-8670, Japan.,Division of Biochemistry &Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chiba 260-8717, Japan.,Saga Medical Centre KOSEIKAN, 400 Nakabaru, Kase-machi, Saga 840-8571, Japan
| |
Collapse
|
60
|
Guan J, Tucker ER, Wan H, Chand D, Danielson LS, Ruuth K, El Wakil A, Witek B, Jamin Y, Umapathy G, Robinson SP, Johnson TW, Smeal T, Martinsson T, Chesler L, Palmer RH, Hallberg B. The ALK inhibitor PF-06463922 is effective as a single agent in neuroblastoma driven by expression of ALK and MYCN. Dis Model Mech 2016; 9:941-52. [PMID: 27483357 PMCID: PMC5047689 DOI: 10.1242/dmm.024448] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 07/04/2016] [Indexed: 12/24/2022] Open
Abstract
The first-in-class inhibitor of ALK, c-MET and ROS1, crizotinib (Xalkori), has shown remarkable clinical efficacy in treatment of ALK-positive non-small cell lung cancer. However, in neuroblastoma, activating mutations in the ALK kinase domain are typically refractory to crizotinib treatment, highlighting the need for more potent inhibitors. The next-generation ALK inhibitor PF-06463922 is predicted to exhibit increased affinity for ALK mutants prevalent in neuroblastoma. We examined PF-06463922 activity in ALK-driven neuroblastoma models in vitro and in vivo In vitro kinase assays and cell-based experiments examining ALK mutations of increasing potency show that PF-06463922 is an effective inhibitor of ALK with greater activity towards ALK neuroblastoma mutants. In contrast to crizotinib, single agent administration of PF-06463922 caused dramatic tumor inhibition in both subcutaneous and orthotopic xenografts as well as a mouse model of high-risk neuroblastoma driven by Th-ALK(F1174L)/MYCN Taken together, our results suggest PF-06463922 is a potent inhibitor of crizotinib-resistant ALK mutations, and highlights an important new treatment option for neuroblastoma patients.
Collapse
Affiliation(s)
- J Guan
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-405 30, Sweden
| | - E R Tucker
- Division of Clinical Studies Cancer Therapeutics, The Institute of Cancer Research, London and Royal Marsden NHS Foundation Trust, Sutton SM2 5NG, UK
| | - H Wan
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-405 30, Sweden
| | - D Chand
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-405 30, Sweden
| | - L S Danielson
- Division of Clinical Studies Cancer Therapeutics, The Institute of Cancer Research, London and Royal Marsden NHS Foundation Trust, Sutton SM2 5NG, UK
| | - K Ruuth
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-405 30, Sweden Department of Molecular Biology, Building 6L, Umeå University, Umeå 901 87, Sweden
| | - A El Wakil
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-405 30, Sweden Department of Molecular Biology, Building 6L, Umeå University, Umeå 901 87, Sweden
| | - B Witek
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-405 30, Sweden Department of Molecular Biology, Building 6L, Umeå University, Umeå 901 87, Sweden
| | - Y Jamin
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London and Royal Marsden NHS Foundation Trust, Sutton SM2 5NG, UK
| | - G Umapathy
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-405 30, Sweden
| | - S P Robinson
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London and Royal Marsden NHS Foundation Trust, Sutton SM2 5NG, UK
| | - T W Johnson
- La Jolla Laboratories, Pfizer Worldwide Research and Development, San Diego, CA 92121, USA
| | - T Smeal
- La Jolla Laboratories, Pfizer Worldwide Research and Development, San Diego, CA 92121, USA
| | - T Martinsson
- Department of Clinical Genetics, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-405 30, Sweden
| | - L Chesler
- Division of Clinical Studies Cancer Therapeutics, The Institute of Cancer Research, London and Royal Marsden NHS Foundation Trust, Sutton SM2 5NG, UK
| | - R H Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-405 30, Sweden
| | - B Hallberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-405 30, Sweden
| |
Collapse
|
61
|
Im JY, Yoon SH, Kim BK, Ban HS, Won KJ, Chung KS, Jung KE, Won M. DNA damage induced apoptosis suppressor (DDIAS) is upregulated via ERK5/MEF2B signaling and promotes β-catenin-mediated invasion. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1449-1458. [PMID: 27412911 DOI: 10.1016/j.bbagrm.2016.07.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 06/06/2016] [Accepted: 07/07/2016] [Indexed: 12/21/2022]
Abstract
DNA damage induced apoptosis suppressor (DDIAS) is an anti-apoptotic protein that promotes cancer cell survival. We previously reported that DDIAS is transcriptionally activated by nuclear factor of activated T cells 2 (NFATc1). However, the upstream regulation of DDIAS expression by growth factors has not been studied. Here, we demonstrate that DDIAS expression is induced by extracellular signal-regulated kinase 5 (ERK5) and myocyte enhancer factor 2B (MEF2B) in response to epidermal growth factor (EGF) and that it positively regulates β-catenin signaling in HeLa cells. The genetic or pharmacological inhibition of ERK5 suppressed DDIAS induction following EGF exposure and the overexpression of constitutively active MEK5 (CA-MEK5) enhanced DDIAS expression. In chromatin immunoprecipitation assays, MEF2B, a downstream target of ERK5, exhibited sequence-specific binding to a MEF2 binding site in the DDIAS promoter following treatment with EGF. The overexpression of MEF2B increased the EGF-mediated induction of DDIAS expression, whereas the knockdown of MEF2B impaired this effect. Furthermore, DDIAS promoted invasion by increasing β-catenin expression at the post-translational level in response to EGF, suggesting that DDIAS plays a crucial role in the metastasis of cancer cells by regulating β-catenin expression. It is unlikely that MEF2B and NFATc1 cooperatively regulate DDIAS transcription in response to EGF. Collectively, EGF activates the ERK5/MEF2 pathway, which in turn induces DDIAS expression to promote cancer cell invasion by activating β-catenin target genes.
Collapse
Affiliation(s)
- Joo-Young Im
- Genomic Personalized Medicine Research Center, KRIBB, Daejeon 305-806, Republic of Korea
| | - Sung-Hoon Yoon
- Genomic Personalized Medicine Research Center, KRIBB, Daejeon 305-806, Republic of Korea; Functional Genomics, University of Science and Technology, Daejeon 305-701, Republic of Korea
| | - Bo-Kyung Kim
- Genomic Personalized Medicine Research Center, KRIBB, Daejeon 305-806, Republic of Korea
| | - Hyun Seung Ban
- Metabolic Regulation Research Center, KRIBB, Daejeon 305-806, Republic of Korea
| | - Kyoung-Jae Won
- Genomic Personalized Medicine Research Center, KRIBB, Daejeon 305-806, Republic of Korea; Functional Genomics, University of Science and Technology, Daejeon 305-701, Republic of Korea
| | - Kyung-Sook Chung
- Metabolic Regulation Research Center, KRIBB, Daejeon 305-806, Republic of Korea
| | - Kyeong Eun Jung
- ST Pharm. Co., LTD, Sihwa Industrial Complex 1, Kyunggido, 429-848, Republic of Korea
| | - Misun Won
- Genomic Personalized Medicine Research Center, KRIBB, Daejeon 305-806, Republic of Korea; Functional Genomics, University of Science and Technology, Daejeon 305-701, Republic of Korea.
| |
Collapse
|
62
|
Simões AES, Rodrigues CMP, Borralho PM. The MEK5/ERK5 signalling pathway in cancer: a promising novel therapeutic target. Drug Discov Today 2016; 21:1654-1663. [PMID: 27320690 DOI: 10.1016/j.drudis.2016.06.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/18/2016] [Accepted: 06/08/2016] [Indexed: 12/18/2022]
Abstract
Conventional mitogen-activated protein kinase (MAPK) family members are among the most sought-after oncogenic effectors for the development of novel human cancer treatment strategies. MEK5/ERK5 has been the less-studied MAPK subfamily, despite its increasingly demonstrated relevance in the growth, survival, and differentiation of normal cells. MEK5/ERK5 signalling has already been proposed to have pivotal roles in several cancer hallmarks, and to mediate the effects of a range of oncogenes. Accumulating evidence indicates the contribution of MEK5/ERK5 signalling to therapy resistance and the benefits of using MEK5/ERK5 inhibitory strategies in the treatment of human cancer. Here, we explore the major known contributions of MEK5/ERK5 signalling to the onset and progression of several types of cancer, and highlight the potential clinical relevance of targeting MEK5/ERK5 pathways.
Collapse
Affiliation(s)
- André E S Simões
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal.
| | - Pedro M Borralho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal.
| |
Collapse
|
63
|
Henssen A, Althoff K, Odersky A, Beckers A, Koche R, Speleman F, Schäfers S, Bell E, Nortmeyer M, Westermann F, De Preter K, Florin A, Heukamp L, Spruessel A, Astrahanseff K, Lindner S, Sadowski N, Schramm A, Astorgues-Xerri L, Riveiro ME, Eggert A, Cvitkovic E, Schulte JH. Targeting MYCN-Driven Transcription By BET-Bromodomain Inhibition. Clin Cancer Res 2016; 22:2470-81. [PMID: 26631615 DOI: 10.1158/1078-0432.ccr-15-1449] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 11/03/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Targeting BET proteins was previously shown to have specific antitumoral efficacy against MYCN-amplified neuroblastoma. We here assess the therapeutic efficacy of the BET inhibitor, OTX015, in preclinical neuroblastoma models and extend the knowledge on the role of BRD4 in MYCN-driven neuroblastoma. EXPERIMENTAL DESIGN The efficacy of OTX015 was assessed in in vitro and in vivo models of human and murine MYCN-driven neuroblastoma. To study the effects of BET inhibition in the context of high MYCN levels, MYCN was ectopically expressed in human and murine cells. The effect of OTX015 on BRD4-regulated transcriptional pause release was analyzed using BRD4 and H3K27Ac chromatin immunoprecipitation coupled with DNA sequencing (ChIP-Seq) and gene expression analysis in neuroblastoma cells treated with OTX015 compared with vehicle control. RESULTS OTX015 showed therapeutic efficacy against preclinical MYCN-driven neuroblastoma models. Similar to previously described BET inhibitors, concurrent MYCN repression was observed in OTX015-treated samples. Ectopic MYCN expression, however, did not abrogate effects of OTX015, indicating that MYCN repression is not the only target of BET proteins in neuroblastoma. When MYCN was ectopically expressed, BET inhibition still disrupted MYCN target gene transcription without affecting MYCN expression. We found that BRD4 binds to super-enhancers and MYCN target genes, and that OTX015 specifically disrupts BRD4 binding and transcription of these genes. CONCLUSIONS We show that OTX015 is effective against mouse and human MYCN-driven tumor models and that BRD4 not only targets MYCN, but specifically occupies MYCN target gene enhancers as well as other genes associated with super-enhancers. Clin Cancer Res; 22(10); 2470-81. ©2015 AACR.
Collapse
Affiliation(s)
- Anton Henssen
- Molecular Pharmacology & Chemistry Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Essen, Germany.
| | - Kristina Althoff
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Essen, Germany. German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Duesseldorf, Essen, Germany
| | - Andrea Odersky
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Essen, Germany
| | - Anneleen Beckers
- Center of Medical Genetics Ghent (CMGG), Ghent University Hospital, Ghent, Belgium
| | - Richard Koche
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Frank Speleman
- Center of Medical Genetics Ghent (CMGG), Ghent University Hospital, Ghent, Belgium
| | - Simon Schäfers
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Essen, Germany. German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Duesseldorf, Essen, Germany
| | - Emma Bell
- Neuroblastoma Genomics, B087, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Maike Nortmeyer
- Neuroblastoma Genomics, B087, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Westermann
- Neuroblastoma Genomics, B087, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katleen De Preter
- Center of Medical Genetics Ghent (CMGG), Ghent University Hospital, Ghent, Belgium
| | - Alexandra Florin
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Lukas Heukamp
- New Oncology, Köln, Germany. Institut für Hämatopathologie Hamburg, Hamburg, Germany
| | - Annika Spruessel
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Essen, Germany
| | - Kathy Astrahanseff
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sven Lindner
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Essen, Germany
| | - Natalie Sadowski
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Essen, Germany
| | - Alexander Schramm
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Essen, Germany
| | | | | | - Angelika Eggert
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Esteban Cvitkovic
- Oncology Therapeutic Development, Clichy, France. Oncoethix, Lausanne, Switzerland
| | - Johannes H Schulte
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Essen, Germany. German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Duesseldorf, Essen, Germany. Neuroblastoma Genomics, B087, German Cancer Research Center (DKFZ), Heidelberg, Germany. Translational Neuro-Oncology, West German Cancer Center (WTZ), University Hospital Essen, University Duisburg-Essen, Essen, Germany. Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
64
|
Guan J, Umapathy G, Yamazaki Y, Wolfstetter G, Mendoza P, Pfeifer K, Mohammed A, Hugosson F, Zhang H, Hsu AW, Halenbeck R, Hallberg B, Palmer RH. FAM150A and FAM150B are activating ligands for anaplastic lymphoma kinase. eLife 2015; 4:e09811. [PMID: 26418745 PMCID: PMC4658194 DOI: 10.7554/elife.09811] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/28/2015] [Indexed: 01/01/2023] Open
Abstract
Aberrant activation of anaplastic lymphoma kinase (ALK) has been described in a range of human cancers, including non-small cell lung cancer and neuroblastoma (Hallberg and Palmer, 2013). Vertebrate ALK has been considered to be an orphan receptor and the identity of the ALK ligand(s) is a critical issue. Here we show that FAM150A and FAM150B are potent ligands for human ALK that bind to the extracellular domain of ALK and in addition to activation of wild-type ALK are able to drive 'superactivation' of activated ALK mutants from neuroblastoma. In conclusion, our data show that ALK is robustly activated by the FAM150A/B ligands and provide an opportunity to develop ALK-targeted therapies in situations where ALK is overexpressed/activated or mutated in the context of the full length receptor. DOI:http://dx.doi.org/10.7554/eLife.09811.001 Cells have receptor proteins on their surface that enable them to detect changes in their environment and communicate with other cells. Signal molecules bind to a segment of the receptor called the extracellular domain that faces out from the cell. This can result in the activation of another domain in the receptor that is just inside the cell, which, in turn, activates signaling pathways that relay the information around the cell. However, these communication systems are often disrupted in cancer cells. This helps the cells to override the strict growth controls imposed upon them by other (healthy) cells in the body. The gene that encodes a receptor protein called Anaplastic Lymphoma Kinase (or ALK for short) is often mutated in some types of human cancer so that the protein is always active. However, we still do not know what signal molecules bind to the ALK protein to activate it in normal cells. Guan, Umapathy et al. used a variety of cell biology and biochemical techniques to study the role of ALK. The experiments show that when either of two proteins called FAM150A and FAM150B are produced in rat nerve cells alongside ALK, the nerve cells rapidly respond and form outgrowths. Experiments using cancer cells derived from human nerve cells also yielded similar results. Guan, Umapathy et al. found that the extracellular domain of ALK can physically interact with FAM150A and FAM150B. The eyes of fruit flies that had been genetically modified to produce the human ALK protein alongside either FAM150A or FAM150B grew more than normal, giving the eyes an abnormal "rough" appearance. Further experiments showed that FAM150A and FAM150B are also able to increase the level of activation of an ALK mutant protein that is already active. Therefore, in future, the development of drugs that stop FAM150A and FAM150B from binding to ALK may be useful for treating cancers that are driven by high levels of ALK activity. Many challenging questions lie ahead to better understand how FAM150A and FAM150B interact with ALK. DOI:http://dx.doi.org/10.7554/eLife.09811.002
Collapse
Affiliation(s)
- Jikui Guan
- Department of Medical Biochemistry and Cell Biology, Instititute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ganesh Umapathy
- Department of Medical Biochemistry and Cell Biology, Instititute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Yasuo Yamazaki
- Department of Medical Biochemistry and Cell Biology, Instititute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Georg Wolfstetter
- Department of Medical Biochemistry and Cell Biology, Instititute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Patricia Mendoza
- Department of Medical Biochemistry and Cell Biology, Instititute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kathrin Pfeifer
- Department of Medical Biochemistry and Cell Biology, Instititute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ateequrrahman Mohammed
- Department of Medical Biochemistry and Cell Biology, Instititute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Hugosson
- Department of Medical Biochemistry and Cell Biology, Instititute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hongbing Zhang
- Five Prime Therapeutics Inc., South San Francisco, United States
| | - Amy W Hsu
- Five Prime Therapeutics Inc., South San Francisco, United States
| | - Robert Halenbeck
- Five Prime Therapeutics Inc., South San Francisco, United States
| | - Bengt Hallberg
- Department of Medical Biochemistry and Cell Biology, Instititute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ruth H Palmer
- Department of Medical Biochemistry and Cell Biology, Instititute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
65
|
Tucker ER, Danielson LS, Innocenti P, Chesler L. Tackling Crizotinib Resistance: The Pathway from Drug Discovery to the Pediatric Clinic. Cancer Res 2015; 75:2770-4. [PMID: 26122839 PMCID: PMC4539575 DOI: 10.1158/0008-5472.can-14-3817] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 03/24/2015] [Indexed: 01/23/2023]
Abstract
Neuroblastoma is a childhood malignancy that has not yet benefitted from the rapid progress in the development of small-molecule therapeutics for cancer. An opportunity to take advantage of pharmaceutical innovation in this area arose when the identification of ALK fusion proteins in non-small cell lung cancer (NSCLC) occurred in parallel to the discovery of point mutations of ALK in neuroblastomas. ALK is now known to be a marker of poor outcome in neuroblastoma, and therefore, urgent development of specific ALK inhibitors to treat this devastating disease is a necessity. However, the translation of small molecules from adult directly into pediatric practice has thus far been challenging, due to mutation-specific structural variances in the ALK kinase domain. We discuss how the most recent structural and biological characterizations of ALK are directing preclinical and clinical studies of ALK inhibitors for both NSCLC and neuroblastoma.
Collapse
Affiliation(s)
- Elizabeth R Tucker
- Paediatric Solid Tumour Biology and Therapeutics Team, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Laura S Danielson
- Paediatric Solid Tumour Biology and Therapeutics Team, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Paolo Innocenti
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Louis Chesler
- Paediatric Solid Tumour Biology and Therapeutics Team, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom.
| |
Collapse
|
66
|
Zhang Y, Li H, Pu Y, Gong S, Liu C, Jiang X, Tao J. Melatonin-mediated inhibition of Purkinje neuron P-type Ca²⁺ channels in vitro induces neuronal hyperexcitability through the phosphatidylinositol 3-kinase-dependent protein kinase C delta pathway. J Pineal Res 2015; 58:321-34. [PMID: 25707622 DOI: 10.1111/jpi.12218] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/20/2015] [Indexed: 12/18/2022]
Abstract
Although melatonin receptors are widely expressed in the mammalian central nervous system and peripheral tissues, there are limited data regarding the functions of melatonin in cerebellar Purkinje cells. Here, we identified a novel functional role of melatonin in modulating P-type Ca(2+) channels and action-potential firing in rat Purkinje neurons. Melatonin at 0.1 μm reversibly decreased peak currents (I(Ba)) by 32.9%. This effect was melatonin receptor 1 (MT(R1)) dependent and was associated with a hyperpolarizing shift in the voltage dependence of inactivation. Pertussis toxin pretreatment, intracellular application of QEHA peptide, and a selective antibody raised against the Gβ subunit prevented the inhibitory effects of melatonin. Pretreatment with phosphatidylinositol 3-kinase (PI3K) inhibitors abolished the melatonin-induced decrease in I(Ba). Surprisingly, melatonin responses were not regulated by Akt, a common downstream target of PI3K. Melatonin treatment significantly increased protein kinase C (PKC) activity 2.1-fold. Antagonists of PKC, but not of protein kinase A, abolished the melatonin-induced decrease in I(Ba). Melatonin application increased the membrane abundance of PKCδ, and PKCδ inhibition (either pharmacologically or genetically) abolished the melatonin-induced IBa response. Functionally, melatonin increased spontaneous action-potential firing by 53.0%; knockdown of MT(R1) and blockade of P-type channels abolished this effect. Thus, our results suggest that melatonin inhibits P-type channels through MT(R1) activation, which is coupled sequentially to the βγ subunits of G(i/o)-protein and to downstream PI3K-dependent PKCδ signaling. This likely contributes to its physiological functions, including spontaneous firing of cerebellar Purkinje neurons.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Neurobiology, Medical College of Soochow University, Suzhou, China; Department of Geriatrics and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | | | | | | | | | | | | |
Collapse
|
67
|
Lindner S, Henssen A, Astrahantseff K, Schulte JH. ALK pERKs up MYCN in neuroblastoma. Sci Signal 2014; 7:pe27. [PMID: 25351246 DOI: 10.1126/scisignal.2005940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The gene expressing the receptor tyrosine kinase anaplastic lymphoma kinase (ALK) is mutated and aberrantly expressed in several cancers. The clinical efficacy of the ALK inhibitor, crizotinib, lags behind expectations for treating MYCN-amplified, ALK-mutant neuroblastoma, a deadly childhood cancer. In this issue of Science Signaling, Umapathy et al. identify the kinase extracellular signal-regulated kinase 5 (ERK5) as a central mediator that enables ALK to boost MYCN expression, and they show that inhibiting ERK5 in concert with ALK reduced neuroblastoma cell viability in vitro and in xenograft tumor models. This report has important clinical implications for the treatment of patients with neuroblastoma or other tumors that overexpress MYC(N) and harbor ALK mutations, such as non-small-cell lung cancer.
Collapse
Affiliation(s)
- Sven Lindner
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, 45122 Essen, Germany. German Cancer Consortium (DKTK), Partner Site Essen/Duesseldorf, 45122 Essen, Germany. Department of Translational Neuro-Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Hufelandstr.55, 45122 Essen, Germany. German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany. Centre for Medical Biotechnology, University Duisburg-Essen, 45122 Essen, Germany
| | - Anton Henssen
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, 45122 Essen, Germany
| | - Kathy Astrahantseff
- Department of Pediatric Oncology, Hematology and Blood and Marrow Transplantation, Charité University Medicine, Campus Virchow Klinikum, 13353 Berlin, Germany
| | - Johannes H Schulte
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, 45122 Essen, Germany. German Cancer Consortium (DKTK), Partner Site Essen/Duesseldorf, 45122 Essen, Germany. Department of Translational Neuro-Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Hufelandstr.55, 45122 Essen, Germany. German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany. Centre for Medical Biotechnology, University Duisburg-Essen, 45122 Essen, Germany.
| |
Collapse
|