Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nature ; 526(7574): 583-6, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26466569

ABSTRACT

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.


Subject(s)
MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Cell Line, Tumor , Enzyme Activation/drug effects , Female , Genes, ras/genetics , Heterocyclic Compounds, 2-Ring/adverse effects , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Indoles/adverse effects , Indoles/pharmacology , MAP Kinase Signaling System/genetics , Mice , Models, Biological , Mutation/genetics , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/adverse effects , Sulfonamides/pharmacology , Vemurafenib
2.
N Engl J Med ; 373(5): 428-37, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26222558

ABSTRACT

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.).


Subject(s)
Aminopyridines/administration & dosage , Giant Cell Tumors/drug therapy , Pyrroles/administration & dosage , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Soft Tissue Neoplasms/drug therapy , Adult , Aged , Aminopyridines/adverse effects , Aminopyridines/pharmacokinetics , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Discovery , Female , Giant Cell Tumors/pathology , Humans , Male , Middle Aged , Pyrroles/adverse effects , Pyrroles/pharmacokinetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Soft Tissue Neoplasms/pathology , Tendons/pathology , Tumor Burden
3.
Proc Natl Acad Sci U S A ; 110(14): 5689-94, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23493555

ABSTRACT

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.


Subject(s)
Aminopyridines/pharmacology , Inflammation/drug therapy , Models, Molecular , Neoplasm Metastasis/drug therapy , Oncogene Protein gp140(v-fms)/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Pyrroles/pharmacology , Aminopyridines/chemical synthesis , Aminopyridines/chemistry , Animals , Cell Survival/drug effects , Chromatography, Affinity , Crystallization , Escherichia coli , Human Umbilical Vein Endothelial Cells , Humans , Indoles , Macrophages/drug effects , Mast Cells/drug effects , Molecular Structure , Mutation, Missense/genetics , Oncogene Protein gp140(v-fms)/chemistry , Oncogene Protein gp140(v-fms)/genetics , Osteoclasts/drug effects , Protein Conformation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-kit/chemistry , Proto-Oncogene Proteins c-kit/genetics , Pyrroles/chemical synthesis , Pyrroles/chemistry , Sf9 Cells , Spodoptera
4.
Nat Rev Drug Discov ; 11(11): 873-86, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23060265

ABSTRACT

The identification of driver oncogenes has provided important targets for drugs that can change the landscape of cancer therapies. One such example is the BRAF oncogene, which is found in about half of all melanomas as well as several other cancers. As a druggable kinase, oncogenic BRAF has become a crucial target of small-molecule drug discovery efforts. Following a rapid clinical development path, vemurafenib (Zelboraf; Plexxikon/Roche) was approved for the treatment of BRAF-mutated metastatic melanoma in the United States in August 2011 and the European Union in February 2012. This Review describes the underlying biology of BRAF, the technology used to identify vemurafenib and its clinical development milestones, along with future prospects based on lessons learned during its development.


Subject(s)
Antineoplastic Agents/therapeutic use , Indoles/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Sulfonamides/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Drug Approval , Drug Design , European Union , Humans , Indoles/pharmacology , Melanoma/genetics , Melanoma/pathology , Mutation , Neoplasm Metastasis , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Sulfonamides/pharmacology , United States , Vemurafenib
5.
Nephrol Dial Transplant ; 26(11): 3458-65, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21804086

ABSTRACT

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is a common cause of renal failure. Aberrant epithelial cell proliferation is a major cause of progressive cyst enlargement in ADPKD. Since activation of the Ras/Raf signaling system has been detected in cyst-lining epithelia, inhibition of Raf kinase has been proposed as an approach to retard the progression of ADPKD. Methods and results. PLX5568, a novel selective small molecule inhibitor of Raf kinases, attenuated proliferation of human ADPKD cyst epithelial cells. It reduced in vitro cyst growth of Madin-Darby Canine Kidney cells and of human ADPKD cells within a collagen gel. In male cy/+ rats with polycystic kidneys, PLX5568 inhibited renal cyst growth along with a significant reduction in the number of proliferating cell nuclear antigen- and phosphorylated extracellular signal-regulated kinase-positive cyst-lining epithelial cells. Furthermore, treated animals showed increased capacity to concentrate urine. However, PLX5568 did not lead to a consistent improvement of renal function. Moreover, although relative cyst volume was decreased, total kidney-to-body weight ratio was not significantly reduced by PLX5568. Further analyses revealed a 2-fold increase of renal and hepatic fibrosis in animals treated with PLX5568. CONCLUSIONS: PLX5568 attenuated cyst enlargement in vitro and in a rat model of ADPKD without improving kidney function, presumably due to increased renal fibrosis. These data suggest that effective therapies for the treatment of ADPKD will need to target fibrosis as well as the growth of cysts.


Subject(s)
Cell Proliferation/drug effects , Cysts/pathology , Kidney/physiopathology , Liver Cirrhosis/chemically induced , Polycystic Kidney Diseases/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Cells, Cultured , Cysts/drug therapy , Dogs , Epithelial Cells/drug effects , Extracellular Signal-Regulated MAP Kinases , Humans , Immunoblotting , Immunoenzyme Techniques , Kidney/drug effects , Male , Mitogen-Activated Protein Kinases , Phosphorylation/drug effects , Polycystic Kidney Diseases/enzymology , Polycystic Kidney Diseases/pathology , Protein Kinase Inhibitors/toxicity , Rats , Rats, Sprague-Dawley
6.
Nature ; 467(7315): 596-9, 2010 Sep 30.
Article in English | MEDLINE | ID: mdl-20823850

ABSTRACT

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.


Subject(s)
Indoles/therapeutic use , Melanoma/drug therapy , Melanoma/enzymology , Mutation/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/therapeutic use , Alleles , Animals , Dogs , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Indoles/administration & dosage , Indoles/adverse effects , Indoles/chemistry , MAP Kinase Signaling System/drug effects , Macaca fascicularis , Melanoma/genetics , Melanoma/pathology , Models, Molecular , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neoplasm Metastasis , Phosphorylation/drug effects , Positron-Emission Tomography , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Rats , Substrate Specificity , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Sulfonamides/chemistry , Vemurafenib , Xenograft Model Antitumor Assays
7.
Neoplasia ; 12(8): 637-49, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20689758

ABSTRACT

PURPOSE: About 65% to 70% of melanomas harbor a mutation in v-raf murine sarcoma viral oncogene homolog B1 (BRAF) that causes the steady-state activation of extracellular signal-regulated kinase (ERK). We sought to investigate the efficacy of PLX4032 (BRAF inhibitor) to identify patterns/predictors of response/resistance and to study the effects of BRAF in melanoma. EXPERIMENTAL DESIGN: Well-characterized melanoma cell lines, including several with acquired drug resistance, were exposed to PLX4032. Growth inhibition, phosphosignaling, cell cycle, apoptosis, and gene expression analyses were performed before and after exposure to drug. RESULTS: Using a growth-adjusted inhibitory concentration of 50% cutoff of 1 microM, 13 of 35 cell lines were sensitive to PLX4032, 16 resistant, and 6 intermediate (37%, 46%, and 17% respectively). PLX4032 caused growth inhibition, G(0)/G(1) arrest, and restored apoptosis in the sensitive cell lines. A BRAF mutation predicted for but did not guarantee a response, whereas a neuroblastoma RAS viral oncogene homolog mutation or wild-type BRAF conferred resistance. Cells with concurrent BRAF mutations and melanocortin 1 receptor germ line variants and/or a more differentiated melanocyte genotype had a preferential response. Acquired PLX4032 resistance reestablishes ERK signaling, promotes a nonmelanocytic genotype, and is associated with an increase in the gene expression of certain metallothioneins and mediators of angiogenesis. CONCLUSIONS: PLX4032 has robust activity in BRAF mutated melanoma. The preclinical use of this molecule identifies criteria for its proper clinical application, describes patterns of and reasons for response/resistance, and affords insight into the role of a BRAF mutation in melanoma.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Indoles/pharmacokinetics , Melanoma/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/metabolism , Sulfonamides/pharmacokinetics , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers, Pharmacological/analysis , Biomarkers, Pharmacological/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , DNA Mutational Analysis , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/pharmacology , Melanoma/genetics , Melanoma/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Substrate Specificity/drug effects , Sulfonamides/pharmacology , Treatment Outcome , Vemurafenib
8.
Proc Natl Acad Sci U S A ; 105(8): 3041-6, 2008 Feb 26.
Article in English | MEDLINE | ID: mdl-18287029

ABSTRACT

BRAF(V600E) 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-Raf(V600E) with an IC(50) of 13 nM, defines a class of kinase inhibitor with marked selectivity in both biochemical and cellular assays. PLX4720 preferentially inhibits the active B-Raf(V600E) 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-Raf(V600E)-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-Raf(V600E)-positive cells. In B-Raf(V600E)-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-Raf(V600E)-driven tumors.


Subject(s)
Apoptosis/drug effects , Indoles/chemistry , Melanoma/drug therapy , Models, Molecular , Oncogenes/genetics , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/chemistry , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Escherichia coli , Humans , Indoles/therapeutic use , Inhibitory Concentration 50 , Mice , Mice, SCID , Molecular Structure , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/therapeutic use
10.
Can J Physiol Pharmacol ; 80(4): 269-74, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12025960

ABSTRACT

Chronically hypoxic rats (exposed to 5000 m elevation for 3 weeks) develop pulmonary hypertension (PH) that is reversed upon return to normoxia and is blocked by bradykinin (BK) antagonist B9430 treatment (100 microg/kg s.c. three times per week). Treatment of rats with both the synthetic VEGF receptor-1/2 antagonist 3-[(2,4-dimethylpyrrol-5-yl)methylidenyl]-indolin-2-one (SU5416) (200 mg/kg, single s.c. injection) and hypoxia (3 weeks) causes irreversible severe PH characterized by marked elevation of pulmonary artery pressure (PAP), right ventricular hypertrophy, and obliteration of pulmonary arteries by proliferating endothelial cells (EC). Between weeks I and 2 of treatment, there is increased apoptotic EC death and caspase-3 activity. The combination of hypoxia with VEGFR-1 and -2 blockade appears to cause death of normal lung EC and proliferation of an apoptosis-resistant proliferating EC phenotype. Cotreatment with BK antagonist B9430 and (or) the broad caspase inhibitor Z-Asp-2,6-dichlorobenzoyloxymethylketone (Z-Asp) (2 mg/kg three times per week) prevented development of severe PH and caused significant reduction of PAP: 39.7 +/- 4.6 mmHg in Z-Asp + SU5416, 37.1 +/- 1.2 mmHg in BK antagonist B9430 + SU5416, 27.2 +/- 0.7 mmHg in Z-Asp alone, and 36.6 +/- 3.0 mmHg in BK antagonist alone versus 48 +/- 1.7 mmHg in SU5416-treated rats and 32.8 +/-1.4 mmHg in vehicle-treated controls. The PAP correlated with the right ventricular mass. Pulmonary arteries of rats treated with Z-Asp and BK antagonist B9430 had a marked reduction of intravascular EC, yet there was still evidence of medial muscular hypertrophy, similar to that observed in chronically hypoxic rats not treated with SU5416. We conclude that EC death induced by VEGFR-2 blockade with SU5416 may trigger an EC selection process that allows for the expansion of apoptosis-resistant EC, possibly driven by mechanisms independent of VEGF and VEGFR-2.


Subject(s)
Bradykinin Receptor Antagonists , Caspase Inhibitors , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Hypertension, Pulmonary/prevention & control , Animals , Caspases/physiology , Enzyme Inhibitors/pharmacology , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/enzymology , Hypoxia/drug therapy , Hypoxia/enzymology , Male , Rats , Rats, Sprague-Dawley , Receptors, Bradykinin/physiology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...