Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Br J Clin Pharmacol ; 82(2): 561, 2016 08.
Article in English | MEDLINE | ID: mdl-27207689

Subject(s)
Disasters , Humans
2.
Neuro Oncol ; 18(4): 557-64, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26449250

ABSTRACT

BACKGROUND: The colony stimulating factor 1 receptor (CSF1R) ligands, CSF1 and interleukin-34, and the KIT ligand, stem cell factor, are expressed in glioblastoma (GB). Microglia, macrophages, blood vessels, and tumor cells also express CSF1R, and depletion of the microglia reduces tumor burden and invasive capacity. PLX3397 is an oral, small molecule that selectively inhibits CSF1R and KIT, penetrates the blood-brain barrier in model systems, and represents a novel approach for clinical development. METHODS: We conducted a phase II study in patients with recurrent GB. The primary endpoint was 6-month progression-free survival (PFS6). Secondary endpoints included overall survival response rate, safety, and plasma/tumor tissue pharmacokinetics. Exploratory endpoints included pharmacodynamic measures of drug effect in blood and tumor tissue. RESULTS: A total of 37 patients were enrolled, with 13 treated prior to a planned surgical resection (Cohort 1) and 24 treated without surgery (Cohort 2). PLX3397 was given at an oral dose of 1000 mg daily and was well tolerated. The primary efficacy endpoint of PFS6 was only 8.6%, with no objective responses. Pharmacokinetic endpoints revealed a median maximal concentration (Cmax) of 8090 ng/mL, with a time to attain Cmax of 2 hour in plasma. Tumor tissue obtained after 7 days of drug exposure revealed a median drug level of 5500 ng/g. Pharmacodynamic changes included an increase in colony stimulating factor 1 and reduced CD14(dim)/CD16+ monocytes in plasma compared with pretreatment baseline values. CONCLUSION: PLX3397 was well tolerated and readily crossed the blood-tumor barrier but showed no efficacy. Additional studies are ongoing, testing combination strategies and potential biomarkers to identify patients with greater likelihood of response.


Subject(s)
Aminopyridines/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Pyrroles/therapeutic use , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Administration, Oral , Aminopyridines/pharmacokinetics , Biomarkers, Tumor/metabolism , Blood-Brain Barrier/drug effects , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cohort Studies , Female , Follow-Up Studies , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , Pyrroles/pharmacokinetics , Tissue Distribution , Tumor Burden
3.
J Clin Oncol ; 33(34): 4032-8, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26460303

ABSTRACT

PURPOSE: BRAF V600E mutation is seen in 5% to 8% of patients with metastatic colorectal cancer (CRC) and is associated with poor prognosis. Vemurafenib, an oral BRAF V600 inhibitor, has pronounced activity in patients with metastatic melanoma, but its activity in patients with BRAF V600E-positive metastatic CRC was unknown. PATIENTS AND METHODS: In this multi-institutional, open-label study, patients with metastatic CRC with BRAF V600 mutations were recruited to an expansion cohort at the previously determined maximum-tolerated dose of 960 mg orally twice a day. RESULTS: Twenty-one patients were enrolled, of whom 20 had received at least one prior metastatic chemotherapy regimen. Grade 3 toxicities included keratoacanthomas, rash, fatigue, and arthralgia. Of the 21 patients treated, one patient had a confirmed partial response (5%; 95% CI, 1% to 24%) and seven other patients had stable disease by RECIST criteria. Median progression-free survival was 2.1 months. Patterns of concurrent mutations, microsatellite instability status, CpG island methylation status, PTEN loss, EGFR expression, and copy number alterations were not associated with clinical benefit. In contrast to prior expectations, concurrent KRAS and NRAS mutations were detected at low allele frequency in a subset of the patients' tumors (median, 0.21% allele frequency) and were apparent mechanisms of acquired resistance in vemurafenib-sensitive patient-derived xenograft models. CONCLUSION: In marked contrast to the results seen in patients with BRAF V600E-mutant melanoma, single-agent vemurafenib did not show meaningful clinical activity in patients with BRAF V600E mutant CRC. Combination strategies are now under development and may be informed by the presence of intratumor heterogeneity of KRAS and NRAS mutations.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Colorectal Neoplasms/drug therapy , Indoles/therapeutic use , Mutation/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/drug therapy , Sulfonamides/therapeutic use , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/secondary , Cohort Studies , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Female , Follow-Up Studies , Humans , Indoles/pharmacokinetics , Male , Middle Aged , Neoplasm Staging , Pilot Projects , Prognosis , Skin Neoplasms/genetics , Skin Neoplasms/mortality , Skin Neoplasms/secondary , Sulfonamides/pharmacokinetics , Survival Rate , Tissue Distribution , Vemurafenib
4.
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
5.
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
6.
Eur J Cancer ; 51(11): 1435-43, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25980594

ABSTRACT

INTRODUCTION: Vemurafenib induces tumour regression in most patients with BRAF(V600E)-mutant melanoma; eventually, most experience progressive disease (PD). Long-term follow-up of patients with BRAF(V600E) melanoma treated in the phase 1 vemurafenib trial is reported. METHODS: Patients received vemurafenib 240-1120 mg (dose escalation cohort) or 960 mg (extension cohort) orally twice daily. Clinical response was evaluated every 8 weeks by Response Evaluation Criteria In Solid Tumors (RECIST). Patients with PD amenable to local therapy (surgery or radiotherapy) were allowed to continue vemurafenib after progression. Overall survival (OS) from time of treatment initiation and from PD was estimated. Sites of PD were recorded. RESULTS: Forty-eight patients (escalation cohort, n = 16; extension cohort, n = 32) received therapeutic doses of vemurafenib (⩾ 240 mg twice daily). Forty-four patients had PD by the time of this analysis and four remained progression free (follow-up time, 1.2-56.1 months). Median OS was 14 months (range, 1.2-56.1); 3- and 4-year melanoma-specific survival rate in the extension cohort was 26% and 19%, respectively. Median OS was 26.0 months (range, 7.7-56.1) among 20 patients who continued vemurafenib after local therapy. Median treatment duration beyond initial PD was 3.8 months (range, 1.1-26.6). In the extension cohort, six and five patients were alive after 3 and 4 years, respectively, on vemurafenib monotherapy. CONCLUSIONS: Some patients with melanoma achieved long-term survival with vemurafenib monotherapy. Continuation of vemurafenib after PD might be beneficial in some patients because remaining disease might continue to respond to BRAF inhibition.


Subject(s)
Indoles/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Sulfonamides/therapeutic use , Adult , Aged , Aged, 80 and over , Disease Progression , Disease-Free Survival , Dose-Response Relationship, Drug , Female , Humans , Male , Melanoma/enzymology , Melanoma/genetics , Melanoma/pathology , Middle Aged , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Survival Rate , Vemurafenib , Young Adult
7.
Oncologist ; 18(3): 314-22, 2013.
Article in English | MEDLINE | ID: mdl-23457002

ABSTRACT

BACKGROUND: Vemurafenib has been approved for the treatment of patients with advanced BRAF(V600E)-mutant melanoma. This report by the Vemurafenib Dermatology Working Group presents the characteristics of dermatologic adverse events (AEs) that occur in vemurafenib-treated patients, including cutaneous squamous cell carcinoma (cuSCC). METHODS: Dermatologic AEs were assessed from three ongoing trials of BRAF(V600E) mutation-positive advanced melanoma. Histologic central review and genetic characterization were completed for a subset of cuSCC lesions. RESULTS: A total of 520 patients received vemurafenib. The most commonly reported AEs were dermatologic AEs, occurring in 92%-95% of patients. Rash was the most common AE (64%-75% of patients), and the most common types were rash not otherwise specified, erythema, maculopapular rash, and folliculitis. Rash development did not appear to correlate with tumor response. Photosensitivity occurred in 35%-63% of patients, and palmar-plantar erythrodysesthesia (PPE) occurred in 8%-10% of patients. The severity of rash, photosensitivity, and PPE were mainly grade 1 or 2. In all, 19%-26% of patients developed cuSCC, mostly keratoacanthomas (KAs). The majority of patients with cuSCC continued therapy without dose reduction after resection. Genetic analysis of 29 cuSCC/KA samples demonstrated HRAS mutations in 41%. CONCLUSIONS: Dermatologic AEs associated with vemurafenib treatment in patients with melanoma were generally manageable with supportive care measures. Dose interruptions and/or reductions were required in <10% of patients.


Subject(s)
Indoles/adverse effects , Indoles/therapeutic use , Melanoma/drug therapy , Neoplasms, Second Primary/chemically induced , Skin Diseases/chemically induced , Skin Neoplasms/drug therapy , Sulfonamides/adverse effects , Sulfonamides/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Clinical Trials as Topic , Female , Humans , Male , Melanoma/pathology , Middle Aged , Neoplasms, Second Primary/therapy , Randomized Controlled Trials as Topic , Skin Neoplasms/pathology , Treatment Outcome , Vemurafenib , Young Adult
8.
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
9.
Thyroid ; 23(10): 1277-83, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23489023

ABSTRACT

BACKGROUND: Clinical benefit from cytotoxic chemotherapy for metastatic papillary thyroid carcinoma (PTC) is disappointing, and effective therapeutic approaches for these patients are urgently needed. Because kinase-activating mutations in the BRAF proto-oncogene commonly occur in advanced PTC, and inhibition of BRAF(V600E) has shown promising clinical activity in melanoma, BRAF inhibitor therapy may be an effective strategy to treat metastatic PTC. METHODS: The dose escalation portion of a first-in-human, phase I study of vemurafenib, a selective RAF inhibitor, included three patients with metastatic PTC harboring the BRAF(V600E) mutation. Vemurafenib was initially dosed at 240-360 mg twice a day, later escalated to 720 mg twice a day. Response evaluation was performed every 8 weeks per Response Evaluation Criteria in Solid Tumors (RECIST). RESULTS: Among the three patients, one had a confirmed partial response with reduction of pulmonary target lesions by 31%, and the duration of response was 7.6 months before the disease progressed in the lungs and the bones. The time to progression was 11.7 months. The other two patients had stable disease, and the time to progression was 13.2 and 11.4 months, respectively. CONCLUSIONS: Vemurafenib appears to have a promising clinical activity in patients with metastatic PTC, and our data suggest that the BRAF(V600E) mutant kinase is a relevant target for therapy in this patient population. Further investigation of inhibitors of mutated BRAF kinase in patients with PTC in a phase II study is warranted.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Papillary/drug therapy , Indoles/therapeutic use , Mutation , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/therapeutic use , Thyroid Neoplasms/drug therapy , Amino Acid Substitution , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Cancer Care Facilities , Carcinoma/drug therapy , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Carcinoma, Papillary/genetics , Carcinoma, Papillary/pathology , Carcinoma, Papillary/secondary , Cohort Studies , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Female , Follow-Up Studies , Humans , Indoles/administration & dosage , Indoles/adverse effects , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Male , Middle Aged , Molecular Targeted Therapy/adverse effects , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Mas , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Survival Analysis , Thyroid Cancer, Papillary , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Tumor Burden/drug effects , Vemurafenib
10.
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
11.
J Clin Oncol ; 30(14): 1628-34, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22454415

ABSTRACT

PURPOSE: Imaging with [(18)F]fluorodeoxyglucose (FDG) -positron emission tomography (PET) allows early recognition of a response to agents that target key driver mutations in human cancer. We aimed to determine the metabolic response rate to vemurafenib in patients with advanced BRAF-mutant melanoma. PATIENTS AND METHODS: Baseline and day 15 FDG-PET was evaluated in 31 patients with advanced melanoma treated in a phase I study of dose escalation of vemurafenib (PLX06-02), which included four patients treated at subtherapeutic doses and 24 patients treated at 960 mg twice a day, which is the maximum-tolerated dose of vemurafenib. RESULTS: All 27 patients treated at potentially therapeutic levels had at least a partial metabolic response, and three patients achieved a complete metabolic response. In the 27 patients, there was an 80% ± 3% reduction in the maximum standardized uptake value (SUVmax) of target lesions and an 87% ± 3% decrease in the percentage of injected dose (%ID) in all identified disease sites. There was a positive correlation between %ID in all identified disease and target-lesion SUVmax (r(2) = 0.66; P < .001) that indicated a significant homogeneity of the response between lesions in individual patients. Although no relationship was found between the reduction in target lesion SUVmax and best response according to RECIST (Response Evaluation Criteria in Solid Tumors), there was a trend for patients with greater reductions in uptake of FDG to have longer progression-free survival. CONCLUSION: FDG-PET is a useful marker of an early biologic response to vemurafenib. Little variability in PET response was found between lesions in individual patients, which suggested minimal intrapatient molecular heterogeneity. FDG-PET is a useful tool for the evaluation of the biologic impact of inhibiting mutant BRAF and may allow for the more effective development of novel agents.


Subject(s)
Fluorodeoxyglucose F18/metabolism , Indoles/therapeutic use , Melanoma/diagnostic imaging , Melanoma/drug therapy , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/drug therapy , Sulfonamides/therapeutic use , Adult , Aged , Aged, 80 and over , Australia , Biomarkers, Tumor/metabolism , Confidence Intervals , Dose-Response Relationship, Drug , Drug Administration Schedule , Early Diagnosis , Female , Follow-Up Studies , Humans , Indoles/adverse effects , Male , Maximum Tolerated Dose , Melanoma/genetics , Melanoma/mortality , Middle Aged , Monitoring, Physiologic , Mutation , Neoplasm Invasiveness/pathology , Neoplasm Staging , Positron-Emission Tomography/methods , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/genetics , Skin Neoplasms/mortality , Statistics, Nonparametric , Sulfonamides/adverse effects , Survival Analysis , Treatment Outcome , Vemurafenib
12.
N Engl J Med ; 366(8): 707-14, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22356324

ABSTRACT

BACKGROUND: Approximately 50% of melanomas harbor activating (V600) mutations in the serine-threonine protein kinase B-RAF (BRAF). The oral BRAF inhibitor vemurafenib (PLX4032) frequently produced tumor regressions in patients with BRAF V600-mutant metastatic melanoma in a phase 1 trial and improved overall survival in a phase 3 trial. METHODS: We designed a multicenter phase 2 trial of vemurafenib in patients with previously treated BRAF V600-mutant metastatic melanoma to investigate the efficacy of vemurafenib with respect to overall response rate (percentage of treated patients with a tumor response), duration of response, and overall survival. The primary end point was the overall response rate as ascertained by the independent review committee; overall survival was a secondary end point. RESULTS: A total of 132 patients had a median follow-up of 12.9 months (range, 0.6 to 20.1). The confirmed overall response rate was 53% (95% confidence interval [CI], 44 to 62; 6% with a complete response and 47% with a partial response), the median duration of response was 6.7 months (95% CI, 5.6 to 8.6), and the median progression-free survival was 6.8 months (95% CI, 5.6 to 8.1). Primary progression was observed in only 14% of patients. Some patients had a response after receiving vemurafenib for more than 6 months. The median overall survival was 15.9 months (95% CI, 11.6 to 18.3). The most common adverse events were grade 1 or 2 arthralgia, rash, photosensitivity, fatigue, and alopecia. Cutaneous squamous-cell carcinomas (the majority, keratoacanthoma type) were diagnosed in 26% of patients. CONCLUSIONS: Vemurafenib induces clinical responses in more than half of patients with previously treated BRAF V600-mutant metastatic melanoma. In this study with a long follow-up, the median overall survival was approximately 16 months. (Funded by Hoffmann-La Roche; ClinicalTrials.gov number, NCT00949702.).


Subject(s)
Indoles/therapeutic use , Melanoma/drug therapy , Mutation , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/therapeutic use , Adult , Aged , Disease Progression , Female , Humans , Indoles/adverse effects , Kaplan-Meier Estimate , Male , Melanoma/genetics , Melanoma/secondary , Middle Aged , Neoplasm Metastasis/drug therapy , Sulfonamides/adverse effects , Treatment Outcome , Vemurafenib
13.
N Engl J Med ; 366(3): 207-15, 2012 Jan 19.
Article in English | MEDLINE | ID: mdl-22256804

ABSTRACT

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.).


Subject(s)
Carcinoma, Squamous Cell/genetics , Genes, ras , Indoles/therapeutic use , Mutation , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/genetics , Sulfonamides/therapeutic use , Aged , Aged, 80 and over , Animals , Carcinoma, Squamous Cell/drug therapy , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Indoles/administration & dosage , Male , Mice , Middle Aged , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Kinase Inhibitors/administration & dosage , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Sulfonamides/administration & dosage , Vemurafenib
14.
N Engl J Med ; 364(26): 2507-16, 2011 Jun 30.
Article in English | MEDLINE | ID: mdl-21639808

ABSTRACT

BACKGROUND: Phase 1 and 2 clinical trials of the BRAF kinase inhibitor vemurafenib (PLX4032) have shown response rates of more than 50% in patients with metastatic melanoma with the BRAF V600E mutation. METHODS: We conducted a phase 3 randomized clinical trial comparing vemurafenib with dacarbazine in 675 patients with previously untreated, metastatic melanoma with the BRAF V600E mutation. Patients were randomly assigned to receive either vemurafenib (960 mg orally twice daily) or dacarbazine (1000 mg per square meter of body-surface area intravenously every 3 weeks). Coprimary end points were rates of overall and progression-free survival. Secondary end points included the response rate, response duration, and safety. A final analysis was planned after 196 deaths and an interim analysis after 98 deaths. RESULTS: At 6 months, overall survival was 84% (95% confidence interval [CI], 78 to 89) in the vemurafenib group and 64% (95% CI, 56 to 73) in the dacarbazine group. In the interim analysis for overall survival and final analysis for progression-free survival, vemurafenib was associated with a relative reduction of 63% in the risk of death and of 74% in the risk of either death or disease progression, as compared with dacarbazine (P<0.001 for both comparisons). After review of the interim analysis by an independent data and safety monitoring board, crossover from dacarbazine to vemurafenib was recommended. Response rates were 48% for vemurafenib and 5% for dacarbazine. Common adverse events associated with vemurafenib were arthralgia, rash, fatigue, alopecia, keratoacanthoma or squamous-cell carcinoma, photosensitivity, nausea, and diarrhea; 38% of patients required dose modification because of toxic effects. CONCLUSIONS: Vemurafenib produced improved rates of overall and progression-free survival in patients with previously untreated melanoma with the BRAF V600E mutation. (Funded by Hoffmann-La Roche; BRIM-3 ClinicalTrials.gov number, NCT01006980.).


Subject(s)
Antineoplastic Agents/therapeutic use , Dacarbazine/therapeutic use , Indoles/therapeutic use , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Dacarbazine/adverse effects , Disease-Free Survival , Female , Humans , Indoles/adverse effects , Intention to Treat Analysis , Male , Melanoma/genetics , Melanoma/mortality , Middle Aged , Mutation , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/adverse effects , Survival Analysis , Vemurafenib , Young Adult
15.
Int J Cancer ; 128(9): 2075-84, 2011 May 01.
Article in English | MEDLINE | ID: mdl-20635392

ABSTRACT

BRAF(V600E) mutations are found in 10% of colorectal cancers (CRCs). The low frequency of this mutation therefore makes it a challenging target for drug development, unless subsets of patients with higher rates of BRAF(V600E) can be defined. Knowledge of the concordance between primary-metastasis pairs and the impact of BRAF(V600E) on outcome would also assist in optimal drug development. We selected primary CRCs from 525 patients (stages I-IV) evenly matched for age (<70 and ≥70), gender and tumor location (right, left and rectum), and 81 primary-metastasis pairs. BRAF(V600E), KRAS mutation and microsatellite instability (MSI) were determined and correlated with clinical features and patient outcomes. In multivariate analyses, increasing patient age (p = 0.04), female gender (p = 0.0005) and right-sided tumor location (p < 0.0001) were independently associated with BRAF(V600E). The prevalence of BRAF(V600E) was considerably higher in older (age > 70) females with KRAS wild-type right-sided colon cancers (50%) compared to the unselected cohort (10%). BRAF(V600E) was associated with inferior overall survival in metastatic CRC (HR = 2.02; 95% CI 1.26-3.26), particularly evident in patients treated with chemotherapy, and is independent of MSI status. BRAF status was concordant in all primary tumors and matched metastases (79 wild-type pairs and two mutant pairs). Clinicopathological and molecular features can identify CRC patients with a higher prevalence of BRAF(V600E). Patients with BRAF(V600E) wild-type primary tumor do not appear to acquire the mutation in their metastases, and BRAF(V600E) is associated with poorer outcomes in metastatic patients. Our findings are timely and will help inform the rational development of BRAF(V600E) inhibitors in CRC.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Age Factors , Aged , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/mortality , Drug Delivery Systems , Drug Resistance, Neoplasm/genetics , Female , Humans , Kaplan-Meier Estimate , Male , Microsatellite Instability , Middle Aged , Neoplasm Staging , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors , Treatment Outcome
16.
N Engl J Med ; 363(9): 809-19, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20818844

ABSTRACT

BACKGROUND: The identification of somatic mutations in the gene encoding the serine-threonine protein kinase B-RAF (BRAF) in the majority of melanomas offers an opportunity to test oncogene-targeted therapy for this disease. METHODS: We conducted a multicenter, phase 1, dose-escalation trial of PLX4032 (also known as RG7204), an orally available inhibitor of mutated BRAF, followed by an extension phase involving the maximum dose that could be administered without adverse effects (the recommended phase 2 dose). Patients received PLX4032 twice daily until they had disease progression. Pharmacokinetic analysis and tumor-response assessments were conducted in all patients. In selected patients, tumor biopsy was performed before and during treatment to validate BRAF inhibition. RESULTS: A total of 55 patients (49 of whom had melanoma) were enrolled in the dose-escalation phase, and 32 additional patients with metastatic melanoma who had BRAF with the V600E mutation were enrolled in the extension phase. The recommended phase 2 dose was 960 mg twice daily, with increases in the dose limited by grade 2 or 3 rash, fatigue, and arthralgia. In the dose-escalation cohort, among the 16 patients with melanoma whose tumors carried the V600E BRAF mutation and who were receiving 240 mg or more of PLX4032 twice daily, 10 had a partial response and 1 had a complete response. Among the 32 patients in the extension cohort, 24 had a partial response and 2 had a complete response. The estimated median progression-free survival among all patients was more than 7 months. CONCLUSIONS: Treatment of metastatic melanoma with PLX4032 in patients with tumors that carry the V600E BRAF mutation resulted in complete or partial tumor regression in the majority of patients. (Funded by Plexxikon and Roche Pharmaceuticals.)


Subject(s)
Antineoplastic Agents/administration & dosage , Indoles/administration & dosage , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Dose-Response Relationship, Drug , Drug Resistance , Drug Resistance, Neoplasm , Female , Humans , Indoles/adverse effects , Indoles/pharmacokinetics , Male , Melanoma/genetics , Melanoma/pathology , Melanoma/secondary , Middle Aged , Mutation , Sulfonamides/adverse effects , Sulfonamides/pharmacokinetics , Treatment Outcome , Vemurafenib , Young Adult
17.
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
18.
Proc Natl Acad Sci U S A ; 106(1): 262-7, 2009 Jan 06.
Article in English | MEDLINE | ID: mdl-19116277

ABSTRACT

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), PPARalpha, PPARgamma, and PPARdelta. Transcriptional transactivation assays were used to select compounds from this chemical series with a bias toward partial agonism toward PPARgamma, to circumvent the clinically observed side effects of full PPARgamma 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 PPARgamma. Compared with full PPARgamma-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).


Subject(s)
Drug Discovery/methods , Hypoglycemic Agents/therapeutic use , PPAR gamma/agonists , Peroxisome Proliferator-Activated Receptors/agonists , Adipocytes/cytology , Adiponectin/genetics , Animals , Cell Differentiation/drug effects , Cell Line , Diabetes Mellitus, Experimental/drug therapy , Humans , Hypoglycemic Agents/pharmacology , Mice , Obesity/drug therapy , PPAR gamma/genetics , Peroxisome Proliferator-Activated Receptors/genetics , Rats , Transcriptional Activation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...