Network


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

Hotspot


Dive into the research topics where Gaston Habets is active.

Publication


Featured researches published by Gaston Habets.


Nature | 2010

Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF -mutant melanoma

Gideon Bollag; Peter Hirth; James H. Tsai; Jiazhong Zhang; Prabha N. Ibrahim; Hanna Cho; Wayne Spevak; Chao Zhang; Ying Zhang; Gaston Habets; Elizabeth A. Burton; Bernice Wong; Garson Tsang; Brian L. West; Ben Powell; Rafe Shellooe; Adhirai Marimuthu; Hoa Nguyen; Kam Y. J. Zhang; Dean R. Artis; Joseph Schlessinger; Fei Su; Brian Higgins; Raman Mahadevan Iyer; Kurt D'Andrea; Astrid Koehler; Michael Stumm; Paul S. Lin; Richard J. Lee; Joseph F. Grippo

B-RAF is the most frequently mutated protein kinase in human cancers. The finding that oncogenic mutations in BRAF are common in melanoma, followed by the demonstration that these tumours are dependent on the RAF/MEK/ERK pathway, offered hope that inhibition of B-RAF kinase activity could benefit melanoma patients. Herein, we describe the structure-guided discovery of PLX4032 (RG7204), a potent inhibitor of oncogenic B-RAF kinase activity. Preclinical experiments demonstrated that PLX4032 selectively blocked the RAF/MEK/ERK pathway in BRAF mutant cells and caused regression of BRAF mutant xenografts. Toxicology studies confirmed a wide safety margin consistent with the high degree of selectivity, enabling Phase 1 clinical trials using a crystalline formulation of PLX4032 (ref. 5). In a subset of melanoma patients, pathway inhibition was monitored in paired biopsy specimens collected before treatment initiation and following two weeks of treatment. This analysis revealed substantial inhibition of ERK phosphorylation, yet clinical evaluation did not show tumour regressions. At higher drug exposures afforded by a new amorphous drug formulation, greater than 80% inhibition of ERK phosphorylation in the tumours of patients correlated with clinical response. Indeed, the Phase 1 clinical data revealed a remarkably high 81% response rate in metastatic melanoma patients treated at an oral dose of 960 mg twice daily. These data demonstrate that BRAF-mutant melanomas are highly dependent on B-RAF kinase activity.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity.

James H. Tsai; John T. Lee; Weiru Wang; Jiazhong Zhang; Hanna Cho; Shumeye Mamo; Ryan Bremer; Sam Gillette; Jun Kong; Nikolas K. Haass; Katrin Sproesser; Ling Li; Keiran S.M. Smalley; Daniel Fong; Yong-Liang Zhu; Adhirai Marimuthu; Hoa Nguyen; Billy Lam; Jennifer Liu; Ivana Cheung; Julie Rice; Yoshihisa Suzuki; Catherine Luu; Calvin Settachatgul; Rafe Shellooe; John Cantwell; Sung-Hou Kim; Joseph Schlessinger; Kam Y. J. Zhang; Brian L. West

BRAFV600E is the most frequent oncogenic protein kinase mutation known. Furthermore, inhibitors targeting “active” protein kinases have demonstrated significant utility in the therapeutic repertoire against cancer. Therefore, we pursued the development of specific kinase inhibitors targeting B-Raf, and the V600E allele in particular. By using a structure-guided discovery approach, a potent and selective inhibitor of active B-Raf has been discovered. PLX4720, a 7-azaindole derivative that inhibits B-RafV600E with an IC50 of 13 nM, defines a class of kinase inhibitor with marked selectivity in both biochemical and cellular assays. PLX4720 preferentially inhibits the active B-RafV600E kinase compared with a broad spectrum of other kinases, and potent cytotoxic effects are also exclusive to cells bearing the V600E allele. Consistent with the high degree of selectivity, ERK phosphorylation is potently inhibited by PLX4720 in B-RafV600E-bearing tumor cell lines but not in cells lacking oncogenic B-Raf. In melanoma models, PLX4720 induces cell cycle arrest and apoptosis exclusively in B-RafV600E-positive cells. In B-RafV600E-dependent tumor xenograft models, orally dosed PLX4720 causes significant tumor growth delays, including tumor regressions, without evidence of toxicity. The work described here represents the entire discovery process, from initial identification through structural and biological studies in animal models to a promising therapeutic for testing in cancer patients bearing B-RafV600E-driven tumors.


The New England Journal of Medicine | 2012

RAS mutations in cutaneous squamous-cell carcinomas in patients treated with BRAF inhibitors.

Fei Su; Amaya Viros; Carla Milagre; Kerstin Trunzer; Gideon Bollag; Olivia Spleiss; Jorge S. Reis-Filho; Xiangju Kong; Richard C. Koya; Keith T. Flaherty; Paul B. Chapman; Min Jung Kim; Robert Hayward; Matthew Martin; Hong Yang; Qiongqing Wang; Holly Hilton; Julie S. Hang; Johannes Noe; Maryou B. Lambros; Felipe C. Geyer; Nathalie Dhomen; Ion Niculescu-Duvaz; Alfonso Zambon; Dan Niculescu-Duvaz; Natasha Preece; Lidia Robert; Nicholas Otte; Stephen Mok; Damien Kee

BACKGROUND Cutaneous squamous-cell carcinomas and keratoacanthomas are common findings in patients treated with BRAF inhibitors. METHODS We performed a molecular analysis to identify oncogenic mutations (HRAS, KRAS, NRAS, CDKN2A, and TP53) in the lesions from patients treated with the BRAF inhibitor vemurafenib. An analysis of an independent validation set and functional studies with BRAF inhibitors in the presence of the prevalent RAS mutation was also performed. RESULTS Among 21 tumor samples, 13 had RAS mutations (12 in HRAS). In a validation set of 14 samples, 8 had RAS mutations (4 in HRAS). Thus, 60% (21 of 35) of the specimens harbored RAS mutations, the most prevalent being HRAS Q61L. Increased proliferation of HRAS Q61L-mutant cell lines exposed to vemurafenib was associated with mitogen-activated protein kinase (MAPK)-pathway signaling and activation of ERK-mediated transcription. In a mouse model of HRAS Q61L-mediated skin carcinogenesis, the vemurafenib analogue PLX4720 was not an initiator or a promoter of carcinogenesis but accelerated growth of the lesions harboring HRAS mutations, and this growth was blocked by concomitant treatment with a MEK inhibitor. CONCLUSIONS Mutations in RAS, particularly HRAS, are frequent in cutaneous squamous-cell carcinomas and keratoacanthomas that develop in patients treated with vemurafenib. The molecular mechanism is consistent with the paradoxical activation of MAPK signaling and leads to accelerated growth of these lesions. (Funded by Hoffmann-La Roche and others; ClinicalTrials.gov numbers, NCT00405587, NCT00949702, NCT01001299, and NCT01006980.).


Nature | 2015

RAF inhibitors that evade paradoxical MAPK pathway activation

Chao Zhang; Wayne Spevak; Ying Zhang; Elizabeth A. Burton; Yan Ma; Gaston Habets; Jiazhong Zhang; Jack Lin; Todd Ewing; Bernice Matusow; Garson Tsang; Adhirai Marimuthu; Hanna Cho; Guoxian Wu; Weiru Wang; Daniel Fong; Hoa Nguyen; Songyuan Shi; Patrick Womack; Marika Nespi; Rafe Shellooe; Heidi Carias; Ben Powell; Emily Light; Laura Sanftner; Jason Walters; James H. Tsai; Brian L. West; Gary Conard Visor; Hamid Rezaei

Oncogenic activation of BRAF fuels cancer growth by constitutively promoting RAS-independent mitogen-activated protein kinase (MAPK) pathway signalling. Accordingly, RAF inhibitors have brought substantially improved personalized treatment of metastatic melanoma. However, these targeted agents have also revealed an unexpected consequence: stimulated growth of certain cancers. Structurally diverse ATP-competitive RAF inhibitors can either inhibit or paradoxically activate the MAPK pathway, depending whether activation is by BRAF mutation or by an upstream event, such as RAS mutation or receptor tyrosine kinase activation. Here we have identified next-generation RAF inhibitors (dubbed ‘paradox breakers’) that suppress mutant BRAF cells without activating the MAPK pathway in cells bearing upstream activation. In cells that express the same HRAS mutation prevalent in squamous tumours from patients treated with RAF inhibitors, the first-generation RAF inhibitor vemurafenib stimulated in vitro and in vivo growth and induced expression of MAPK pathway response genes; by contrast the paradox breakers PLX7904 and PLX8394 had no effect. Paradox breakers also overcame several known mechanisms of resistance to first-generation RAF inhibitors. Dissociating MAPK pathway inhibition from paradoxical activation might yield both improved safety and more durable efficacy than first-generation RAF inhibitors, a concept currently undergoing human clinical evaluation with PLX8394.


The New England Journal of Medicine | 2015

Structure-Guided Blockade of CSF1R Kinase in Tenosynovial Giant-Cell Tumor

William D. Tap; Zev A. Wainberg; Stephen P. Anthony; Prabha N. Ibrahim; Chao Zhang; John H. Healey; Bartosz Chmielowski; Arthur P. Staddon; Allen Lee Cohn; Geoffrey I. Shapiro; Vicki L. Keedy; Arun S. Singh; Igor Puzanov; Eunice L. Kwak; Andrew J. Wagner; Daniel D. Von Hoff; Glen J. Weiss; Ramesh K. Ramanathan; Jiazhong Zhang; Gaston Habets; Ying Zhang; Elizabeth A. Burton; Gary Conard Visor; Laura Sanftner; Paul Severson; Hoa Nguyen; Marie J. Kim; Adhirai Marimuthu; Garson Tsang; Rafe Shellooe

BACKGROUND Expression of the colony-stimulating factor 1 (CSF1) gene is elevated in most tenosynovial giant-cell tumors. This observation has led to the discovery and clinical development of therapy targeting the CSF1 receptor (CSF1R). METHODS Using x-ray co-crystallography to guide our drug-discovery research, we generated a potent, selective CSF1R inhibitor, PLX3397, that traps the kinase in the autoinhibited conformation. We then conducted a multicenter, phase 1 trial in two parts to analyze this compound. In the first part, we evaluated escalations in the dose of PLX3397 that was administered orally in patients with solid tumors (dose-escalation study). In the second part, we evaluated PLX3397 at the chosen phase 2 dose in an extension cohort of patients with tenosynovial giant-cell tumors (extension study). Pharmacokinetic and tumor responses in the enrolled patients were assessed, and CSF1 in situ hybridization was performed to confirm the mechanism of action of PLX3397 and that the pattern of CSF1 expression was consistent with the pathological features of tenosynovial giant-cell tumor. RESULTS A total of 41 patients were enrolled in the dose-escalation study, and an additional 23 patients were enrolled in the extension study. The chosen phase 2 dose of PLX3397 was 1000 mg per day. In the extension study, 12 patients with tenosynovial giant-cell tumors had a partial response and 7 patients had stable disease. Responses usually occurred within the first 4 months of treatment, and the median duration of response exceeded 8 months. The most common adverse events included fatigue, change in hair color, nausea, dysgeusia, and periorbital edema; adverse events rarely led to discontinuation of treatment. CONCLUSIONS Treatment of tenosynovial giant-cell tumors with PLX3397 resulted in a prolonged regression in tumor volume in most patients. (Funded by Plexxikon; ClinicalTrials.gov number, NCT01004861.).


Proceedings of the National Academy of Sciences of the United States of America | 2009

Scaffold-based discovery of indeglitazar, a PPAR pan-active anti-diabetic agent

Dean R. Artis; J. J. Lin; Chao Zhang; Weiru Wang; U. Mehra; M. Perreault; D. Erbe; H. I. Krupka; B. P. England; J. Arnold; A. N. Plotnikov; Adhirai Marimuthu; Hoa Nguyen; S. Will; M. Signaevsky; J. Kral; J. Cantwell; C. Settachatgull; D. S. Yan; Daniel Fong; A. Oh; S. Shi; P. Womack; Ben Powell; Gaston Habets; Brian L. West; Kam Y. J. Zhang; M. V. Milburn; G. P. Vlasuk; K. P. Hirth

In a search for more effective anti-diabetic treatment, we used a process coupling low-affinity biochemical screening with high-throughput co-crystallography in the design of a series of compounds that selectively modulate the activities of all three peroxisome proliferator-activated receptors (PPARs), PPARα, PPARγ, and PPARδ. Transcriptional transactivation assays were used to select compounds from this chemical series with a bias toward partial agonism toward PPARγ, to circumvent the clinically observed side effects of full PPARγ agonists. Co-crystallographic characterization of the lead molecule, indeglitazar, in complex with each of the 3 PPARs revealed the structural basis for its PPAR pan-activity and its partial agonistic response toward PPARγ. Compared with full PPARγ-agonists, indeglitazar is less potent in promoting adipocyte differentiation and only partially effective in stimulating adiponectin gene expression. Evaluation of the compound in vivo confirmed the reduced adiponectin response in animal models of obesity and diabetes while revealing strong beneficial effects on glucose, triglycerides, cholesterol, body weight, and other metabolic parameters. Indeglitazar has now progressed to Phase II clinical evaluations for Type 2 diabetes mellitus (T2DM).


Proceedings of the National Academy of Sciences of the United States of America | 2013

Design and pharmacology of a highly specific dual FMS and KIT kinase inhibitor

Chao Zhang; Prabha N. Ibrahim; Jiazhong Zhang; Elizabeth A. Burton; Gaston Habets; Ying Zhang; Ben Powell; Brian L. West; Bernice Matusow; Garson Tsang; Rafe Shellooe; Heidi Carias; Hoa Nguyen; Adhirai Marimuthu; Kam Y. J. Zhang; Angela Oh; Ryan Bremer; Clarence R. Hurt; Dean R. Artis; Guoxian Wu; Marika Nespi; Wayne Spevak; Paul S. Lin; Keith Nolop; Peter Hirth; Gregory H Tesch; Gideon Bollag

Inflammation and cancer, two therapeutic areas historically addressed by separate drug discovery efforts, are now coupled in treatment approaches by a growing understanding of the dynamic molecular dialogues between immune and cancer cells. Agents that target specific compartments of the immune system, therefore, not only bring new disease modifying modalities to inflammatory diseases, but also offer a new avenue to cancer therapy by disrupting immune components of the microenvironment that foster tumor growth, progression, immune evasion, and treatment resistance. McDonough feline sarcoma viral (v-fms) oncogene homolog (FMS) and v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog (KIT) are two hematopoietic cell surface receptors that regulate the development and function of macrophages and mast cells, respectively. We disclose a highly specific dual FMS and KIT kinase inhibitor developed from a multifaceted chemical scaffold. As expected, this inhibitor blocks the activation of macrophages, osteoclasts, and mast cells controlled by these two receptors. More importantly, the dual FMS and KIT inhibition profile has translated into a combination of benefits in preclinical disease models of inflammation and cancer.


Nature Biotechnology | 2011

Interrogating the kinome

Chao Zhang; Gaston Habets; Gideon Bollag

Comprehensive studies of the kinome set the stage for discovering the next generation of kinase-directed drugs.


Methods in Enzymology | 2001

cDNA array analyses of K-ras-induced gene transcription

Gaston Habets; Marc Knepper; Jaina Sumortin; Yun-Jung Choi; Takehiko Sasazuki; Senji Shirasawa; Gideon Bollag

Publisher Summary This chapter describes cDNA array analyses of K-Ras-induced gene transcription. Among all oncogene-encoded proteins, the Ras proteins are probably the best studied with regard to their biochemical mechanism and how the mutations that are found in human tumors affect their activity. To characterize the quantitative and qualitative differences in gene expression during tumor transformation, the chapter describes the conditional reconstitution of the expression of an activated K- ras allele in the K-RasD13 knockout cell clones DKO-4 and Hke-3.The chapter presents a set of experiments designed to identify transcripts that respond to K-Ras activity. These experiments were conducted in cell lines whose morphological transformation had been reverted by deletion of an activated K-ras allele. Biochemical analysis of these K-Ras expressing cells confirms activation of K-Ras pathways and accompanying gene expression. The methods described here prove useful to unravel K-Ras-dependent transcription. Given the central role of K-Ras in many types of cancer, it is hoped that the identification of K-Ras transcriptional targets will lead to new therapeutic targets, and ultimately to new cancer cures.


Pain | 2015

Targeting cells of the myeloid lineage attenuates pain and disease progression in a prostate model of bone cancer.

Michelle L. Thompson; Juan Miguel Jimenez-Andrade; Stephane R. Chartier; James H. Tsai; Elizabeth A. Burton; Gaston Habets; Paul S. Lin; Brian L. West; Patrick W. Mantyh

Abstract Tumor cells frequently metastasize to bone where they can generate cancer-induced bone pain (CIBP) that can be difficult to fully control using available therapies. Here, we explored whether PLX3397, a high-affinity small molecular antagonist that binds to and inhibits phosphorylation of colony-stimulating factor-1 receptor, the tyrosine-protein kinase c-Kit, and the FMS-like tyrosine kinase 3, can reduce CIBP. These 3 targets all regulate the proliferation and function of a subset of the myeloid cells including macrophages, osteoclasts, and mast cells. Preliminary experiments show that PLX3397 attenuated inflammatory pain after formalin injection into the hind paw of the rat. As there is an inflammatory component in CIBP, involving macrophages and osteoclasts, the effect of PLX3397 was explored in a prostate model of CIBP where skeletal pain, cancer cell proliferation, tumor metastasis, and bone remodeling could be monitored in the same animal. Administration of PLX3397 was initiated on day 14 after prostate cancer cell injection when the tumor was well established, and tumor-induced bone remodeling was first evident. Over the next 6 weeks, sustained administration of PLX3397 attenuated CIBP behaviors by approximately 50% and was equally efficacious in reducing tumor cell growth, formation of new tumor colonies in bone, and pathological tumor-induced bone remodeling. Developing a better understanding of potential effects that analgesic therapies have on the tumor itself may allow the development of therapies that not only better control the pain but also positively impact disease progression and overall survival in patients with bone cancer.

Collaboration


Dive into the Gaston Habets's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge