Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Louis Chesler is active.

Publication


Featured researches published by Louis Chesler.


Journal of Clinical Investigation | 1996

Captopril inhibits angiogenesis and slows the growth of experimental tumors in rats.

Olga V. Volpert; William F. Ward; Mark W. Lingen; Louis Chesler; Dennis B. Solt; Mark D. Johnson; Agostino Molteni; Peter J. Polverini; Noel P. Bouck

Captopril, an inhibitor of angiotensin converting enzyme, is widely used clinically to manage hypertension and congestive heart failure. Here captopril is shown to be an inhibitor of angiogenesis able to block neovascularization induced in the rat cornea. Captopril acted directly and specifically on capillary endothelial cells, inhibiting their chemotaxis with a biphasic dose-response curve showing an initial decrease at clinically achievable doses under 10 microM and a further slow decline in the millimolar range. Captopril inhibition of endothelial cell migration was not mediated by angiotensin converting enzyme inhibition, but was suppressed by zinc. Direct inhibition by captopril of zinc-dependent endothelial cell-derived 72-and 92-kD metalloproteinases known to be essential for angiogenesis was also seen. When used systemically on rats captopril inhibited corneal neovascularization and showed the antitumor activity expected of an inhibitor of angiogenesis, decreasing the number of mitoses present in carcinogen-induced foci of preneoplastic liver cells and slowing the growth rate of an experimental fibrosarcoma whose cells were resistant to captopril in vitro. These data define this widely used drug as a new inhibitor of neovascularization and raise the possibility that patients on long term captopril therapy may derive unexpected benefits from its antiangiogenic activities.


Cancer Cell | 2012

The ALK(F1174L) mutation potentiates the oncogenic activity of MYCN in neuroblastoma.

Teeara Berry; William Luther; Namrata Bhatnagar; Yann Jamin; Evon Poon; Takaomi Sanda; De-Sheng Pei; Bandana Sharma; Winston R. Vetharoy; Albert Hallsworth; Zai Ahmad; Karen Barker; Lisa A. Moreau; Hannah Webber; Wenchao Wang; Qingsong Liu; Antonio R. Perez-Atayde; Scott J. Rodig; Nai-Kong Cheung; Florence I. Raynaud; Bengt Hallberg; Simon P. Robinson; Nathanael S. Gray; Andrew D.J. Pearson; Suzanne A. Eccles; Louis Chesler; Rani E. George

The ALK(F1174L) mutation is associated with intrinsic and acquired resistance to crizotinib and cosegregates with MYCN in neuroblastoma. In this study, we generated a mouse model overexpressing ALK(F1174L) in the neural crest. Compared to ALK(F1174L) and MYCN alone, co-expression of these two oncogenes led to the development of neuroblastomas with earlier onset, higher penetrance, and enhanced lethality. ALK(F1174L)/MYCN tumors exhibited increased MYCN dosage due to ALK(F1174L)-induced activation of the PI3K/AKT/mTOR and MAPK pathways, coupled with suppression of MYCN pro-apoptotic effects. Combined treatment with the ATP-competitive mTOR inhibitor Torin2 overcame the resistance of ALK(F1174L)/MYCN tumors to crizotinib. Our findings demonstrate a pathogenic role for ALK(F1174L) in neuroblastomas overexpressing MYCN and suggest a strategy for improving targeted therapy for ALK-positive neuroblastoma.


Cancer Research | 2006

Inhibition of phosphatidylinositol 3-kinase destabilizes mycn protein and blocks malignant progression in neuroblastoma

Louis Chesler; Chris Schlieve; David D. Goldenberg; Anna Marie Kenney; Grace E. Kim; Alex McMillan; Katherine K. Matthay; David H. Rowitch; William A. Weiss

Amplification of MYCN occurs commonly in neuroblastoma. We report that phosphatidylinositol 3-kinase (PI3K) inhibition in murine neuroblastoma (driven by a tyrosine hydroxylase-MYCN transgene) led to decreased tumor mass and decreased levels of Mycn protein without affecting levels of MYCN mRNA. Consistent with these observations, PI3K inhibition in MYCN-amplified human neuroblastoma cell lines resulted in decreased levels of Mycn protein without affecting levels of MYCN mRNA and caused decreased proliferation and increased apoptosis. To clarify the importance of Mycn as a target of broad-spectrum PI3K inhibitors, we transduced wild-type N-myc and N-myc mutants lacking glycogen synthase kinase 3beta phosphorylation sites into human neuroblastoma cells with no endogenous expression of myc. In contrast to wild-type N-myc, the phosphorylation-defective mutant proteins were stabilized and were resistant to the antiproliferative effects of PI3K inhibition. Our results show the importance of Mycn as a therapeutic target in established tumors in vivo, offer a mechanistic rationale to test PI3K inhibitors in MYCN-amplified neuroblastoma, and represent a therapeutic approach applicable to a broad range of cancers in which transcription factors are stabilized through a PI3K-dependent mechanism.


Genes & Development | 2010

Pleiotropic role for MYCN in medulloblastoma

Fredrik J. Swartling; Matthew R. Grimmer; Christopher S. Hackett; Paul A. Northcott; Qi-Wen Fan; David D. Goldenberg; Jasmine Lau; Selma Masic; Kim Nguyen; Slava Yakovenko; Xiao-Ning Zhe; Heather C. Flynn Gilmer; Rodney Collins; Mai Nagaoka; Joanna J. Phillips; Robert B. Jenkins; Tarik Tihan; Scott R. VandenBerg; C. David James; Kohichi Tanaka; Michael D. Taylor; William A. Weiss; Louis Chesler

Medulloblastoma (MB) is the most common malignant brain tumor of childhood. Sonic Hedgehog (SHH) signaling drives a minority of MB, correlating with desmoplastic pathology and favorable outcome. The majority, however, arises independently of SHH and displays classic or large cell anaplastic (LCA) pathology and poor prognosis. To identify common signaling abnormalities, we profiled mRNA, demonstrating misexpression of MYCN in the majority of human MB and negligible expression in normal cerebella. We clarified a role in pathogenesis by targeting MYCN (and luciferase) to cerebella of transgenic mice. MYCN-driven MB showed either classic or LCA pathologies, with Shh signaling activated in approximately 5% of tumors, demonstrating that MYCN can drive MB independently of Shh. MB arose at high penetrance, consistent with a role for MYCN in initiation. Tumor burden correlated with bioluminescence, with rare metastatic spread to the leptomeninges, suggesting roles for MYCN in both progression and metastasis. Transient pharmacological down-regulation of MYCN led to both clearance and senescence of tumor cells, and improved survival. Targeted expression of MYCN thus contributes to initiation, progression, and maintenance of MB, suggesting a central role for MYCN in pathogenesis.


Cancer Cell | 2012

Distinct Neural Stem Cell Populations Give Rise to Disparate Brain Tumors in Response to N-MYC

Fredrik J. Swartling; Vasil Savov; Anders Persson; Justin Chen; Christopher S. Hackett; Paul A. Northcott; Matthew R. Grimmer; Jasmine Lau; Louis Chesler; Arie Perry; Joanna J. Phillips; Michael D. Taylor; William A. Weiss

The proto-oncogene MYCN is mis-expressed in various types of human brain tumors. To clarify how developmental and regional differences influence transformation, we transduced wild-type or mutationally stabilized murine N-myc(T58A) into neural stem cells (NSCs) from perinatal murine cerebellum, brain stem, and forebrain. Transplantation of N-myc(WT) NSCs was insufficient for tumor formation. N-myc(T58A) cerebellar and brain stem NSCs generated medulloblastoma/primitive neuroectodermal tumors, whereas forebrain NSCs developed diffuse glioma. Expression analyses distinguished tumors generated from these different regions, with tumors from embryonic versus postnatal cerebellar NSCs demonstrating Sonic Hedgehog (SHH) dependence and SHH independence, respectively. These differences were regulated in part by the transcription factor SOX9, activated in the SHH subclass of human medulloblastoma. Our results demonstrate context-dependent transformation of NSCs in response to a common oncogenic signal.


Cancer Cell | 2013

Small Molecule Inhibitors of Aurora-A Induce Proteasomal Degradation of N-Myc in Childhood Neuroblastoma.

Markus Brockmann; Evon Poon; Teeara Berry; Anne Carstensen; Hedwig E. Deubzer; Lukas Rycak; Yann Jamin; Khin Thway; Simon P. Robinson; Frederik Roels; Olaf Witt; Matthias Fischer; Louis Chesler; Martin Eilers

Amplification of MYCN is a driver mutation in a subset of human neuroendocrine tumors, including neuroblastoma. No small molecules that target N-Myc, the protein encoded by MYCN, are clinically available. N-Myc forms a complex with the Aurora-A kinase, which protects N-Myc from proteasomal degradation. Although stabilization of N-Myc does not require the catalytic activity of Aurora-A, we show here that two Aurora-A inhibitors, MLN8054 and MLN8237, disrupt the Aurora-A/N-Myc complex and promote degradation of N-Myc mediated by the Fbxw7 ubiquitin ligase. Disruption of the Aurora-A/N-Myc complex inhibits N-Myc-dependent transcription, correlating with tumor regression and prolonged survival in a mouse model of MYCN-driven neuroblastoma. We conclude that Aurora-A is an accessible target that makes destabilization of N-Myc a viable therapeutic strategy.


Cancer Cell | 2015

Combined MYC and P53 defects emerge at medulloblastoma relapse and define rapidly progressive, therapeutically targetable disease

Rebecca Hill; Sanne Kuijper; Janet C. Lindsey; Kevin Petrie; Ed Schwalbe; Karen Barker; Jessica K.R. Boult; Daniel Williamson; Zai Ahmad; Albert Hallsworth; Sarra L. Ryan; Evon Poon; Simon P. Robinson; Ruth Ruddle; Florence I. Raynaud; Louise Howell; Colin Kwok; Abhijit Joshi; Sl Nicholson; Stephen Crosier; David W. Ellison; Stephen B. Wharton; Keith Robson; Antony Michalski; Darren Hargrave; Ts Jacques; Barry Pizer; Simon Bailey; Fredrik J. Swartling; William A. Weiss

Summary We undertook a comprehensive clinical and biological investigation of serial medulloblastoma biopsies obtained at diagnosis and relapse. Combined MYC family amplifications and P53 pathway defects commonly emerged at relapse, and all patients in this group died of rapidly progressive disease postrelapse. To study this interaction, we investigated a transgenic model of MYCN-driven medulloblastoma and found spontaneous development of Trp53 inactivating mutations. Abrogation of p53 function in this model produced aggressive tumors that mimicked characteristics of relapsed human tumors with combined P53-MYC dysfunction. Restoration of p53 activity and genetic and therapeutic suppression of MYCN all reduced tumor growth and prolonged survival. Our findings identify P53-MYC interactions at medulloblastoma relapse as biomarkers of clinically aggressive disease that may be targeted therapeutically.


Clinical Cancer Research | 2013

New Strategies in Neuroblastoma: Therapeutic Targeting of MYCN and ALK

Giuseppe Barone; John Anderson; Andrew D.J. Pearson; Kevin Petrie; Louis Chesler

Clinical outcome remains poor in patients with high-risk neuroblastoma, in which chemoresistant relapse is common following high-intensity conventional multimodal therapy. Novel treatment approaches are required. Although recent genomic profiling initiatives have not revealed a high frequency of mutations in any significant number of therapeutically targeted genes, two exceptions, amplification of the MYCN oncogene and somatically acquired tyrosine kinase domain point mutations in anaplastic lymphoma kinase (ALK), present exciting possibilities for targeted therapy. In contrast with the situation with ALK, in which a robust pipeline of pharmacologic agents is available from early clinical use in adult malignancy, therapeutic targeting of MYCN (and MYC oncoproteins in general) represents a significant medicinal chemistry challenge that has remained unsolved for two decades. We review the latest approaches envisioned for blockade of ALK activity in neuroblastoma, present a classification of potential approaches for therapeutic targeting of MYCN, and discuss how recent developments in targeting of MYC proteins seem to make therapeutic inhibition of MYCN a reality in the clinic. Clin Cancer Res; 19(21); 5814–21. ©2013 AACR.


Cancer Research | 2007

Malignant Progression and Blockade of Angiogenesis in a Murine Transgenic Model of Neuroblastoma

Louis Chesler; David D. Goldenberg; Isha T. Seales; Ronit Satchi-Fainaro; Matthew R. Grimmer; Rodney Collins; Chris Struett; Kim Nguyen; Grace E. Kim; Tarik Tihan; Yun Bao; Rolf A. Brekken; Gabriele Bergers; Judah Folkman; William A. Weiss

Targeted expression of MYCN to the neural crest [under control of the rat tyrosine hydroxylase (TH) promoter] causes neuroblastoma in transgenic mice (TH-MYCN) and is a well-established model for this disease. Because high levels of MYCN are associated with enhanced tumor angiogenesis and poor clinical outcome in neuroblastoma, we serially characterized malignant progression, angiogenesis, and sensitivity to angiogenic blockade in tumors from these animals. Tumor cells were proliferative, secreted high levels of the angiogenic ligand vascular endothelial growth factor (VEGF), and recruited a complex vasculature expressing the angiogenic markers VEGF-R2, alpha-SMA, and matrix metalloproteinases MMP-2 and MMP-9, all of which are also expressed in human disease. Treatment of established murine tumors with the angiogenesis inhibitor TNP-470 caused near-complete ablation, with reduced proliferation, enhanced apoptosis, and vasculature disruption. Because TNP-470 has been associated with neurotoxicity, we tested the recently described water-soluble HPMA copolymer-TNP-470 conjugate (caplostatin), which showed comparable efficacy and was well tolerated without weight loss or neurotoxicity as measured by rotarod testing. This study highlights the importance of angiogenesis inhibition in a spontaneous murine tumor with native tumor-microenvironment interactions, validates the use of mice transgenic for TH-MYCN as a model for therapy in this common pediatric tumor, and supports further clinical development of caplostatin as an antiangiogenic therapy in childhood neuroblastoma.


Clinical Cancer Research | 2012

CCT244747 is a novel, potent and selective CHK1 inhibitor with oral efficacy alone and in combination with genotoxic anticancer drugs

Mike I. Walton; Paul D. Eve; Angela Hayes; Melanie Valenti; Alexis de Haven Brandon; Gary Box; Albert Hallsworth; Elizabeth L. Smith; Kathy Boxall; Michael Lainchbury; Thomas P. Matthews; Yann Jamin; Simon P. Robinson; G. Wynne Aherne; John C. Reader; Louis Chesler; Florence I. Raynaud; Suzanne A. Eccles; Ian Collins; Michelle D. Garrett

Purpose: Many tumors exhibit defective cell-cycle checkpoint control and increased replicative stress. CHK1 is critically involved in the DNA damage response and maintenance of replication fork stability. We have therefore discovered a novel potent, highly selective, orally active ATP-competitive CHK1 inhibitor, CCT244747, and present its preclinical pharmacology and therapeutic activity. Experimental Design: Cellular CHK1 activity was assessed using an ELISA assay, and cytotoxicity a SRB assay. Biomarker modulation was measured using immunoblotting, and cell-cycle effects by flow cytometry analysis. Single-agent oral CCT244747 antitumor activity was evaluated in a MYCN-driven transgenic mouse model of neuroblastoma by MRI and in genotoxic combinations in human tumor xenografts by growth delay. Results: CCT244747 inhibited cellular CHK1 activity (IC50 29–170 nmol/L), significantly enhanced the cytotoxicity of several anticancer drugs, and abrogated drug-induced S and G2 arrest in multiple tumor cell lines. Biomarkers of CHK1 (pS296 CHK1) activity and cell-cycle inactivity (pY15 CDK1) were induced by genotoxics and inhibited by CCT244747 both in vitro and in vivo, producing enhanced DNA damage and apoptosis. Active tumor concentrations of CCT244747 were obtained following oral administration. The antitumor activity of both gemcitabine and irinotecan were significantly enhanced by CCT244747 in several human tumor xenografts, giving concomitant biomarker modulation indicative of CHK1 inhibition. CCT244747 also showed marked antitumor activity as a single agent in a MYCN-driven neuroblastoma. Conclusion: CCT244747 represents the first structural disclosure of a highly selective, orally active CHK1 inhibitor and warrants further evaluation alone or combined with genotoxic anticancer therapies. Clin Cancer Res; 18(20); 5650–61. ©2012 AACR.

Collaboration


Dive into the Louis Chesler's collaboration.

Top Co-Authors

Avatar

Simon P. Robinson

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Yann Jamin

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Albert Hallsworth

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Andrew D.J. Pearson

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Evon Poon

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Karen Barker

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Hannah Webber

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Jessica K.R. Boult

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Laura Danielson

Institute of Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge