Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
NPJ Precis Oncol ; 6(1): 13, 2022 Mar 01.
Article En | MEDLINE | ID: mdl-35233056

The NCI-MATCH was designed to characterize the efficacy of targeted therapies in histology-agnostic driver mutation-positive malignancies. Sub-protocols F and G were developed to evaluate the role of crizotinib in rare tumors that harbored either ALK or ROS1 rearrangements. Patients with malignancies that progressed following at least one prior systemic therapy were accrued to the NCI-MATCH for molecular profiling, and those with actionable ALK or ROS1 rearrangements were offered participation in sub-protocols F or G, respectively. There were five patients who enrolled on Arm F (ALK) and four patients on Arm G (ROS1). Few grade 3 or 4 toxicities were noted, including liver test abnormalities, and acute kidney injury. For sub-protocol F (ALK), the response rate was 50% (90% CI 9.8-90.2%) with one complete response among the 4 eligible patients. The median PFS was 3.8 months, and median OS was 4.3 months. For sub-protocol G (ROS1) the response rate was 25% (90% CI 1.3-75.1%). The median PFS was 4.3 months, and median OS 6.2 months. Data from 3 commercial vendors showed that the prevalence of ALK and ROS1 rearrangements in histologies other than non-small cell lung cancer and lymphoma was rare (0.1% and 0.4% respectively). We observed responses to crizotinib which met the primary endpoint for ALK fusions, albeit in a small number of patients. Despite the limited accrual, some of the patients with these oncogenic fusions can respond to crizotinib which may have a therapeutic role in this setting.

3.
Br J Dermatol ; 185(2): 253-262, 2021 08.
Article En | MEDLINE | ID: mdl-32652567

The therapeutic landscape for melanoma has evolved drastically in the past decade. Currently, immune checkpoint inhibitors and small-molecule inhibitors targeting the mitogen-activated protein kinase (MAPK) pathway are the two mainstay therapies for BRAFV600 mutant advanced melanoma. Although MAPK dependence has been variably demonstrated in melanomas lacking BRAFV600 mutations, definitive evidence of benefit with MAPK inhibitors has not been demonstrated. Thus, in the BRAFV600 'wild-type' setting, immune checkpoint inhibitors are the standalone option(s). In the BRAFV600 mutant setting, there is no definitive evidence prioritizing one therapeutic modality over another. Herein, we review the updated data of the pivotal phase III randomized controlled trials that established the standard-of-care first-line treatment for advanced melanoma, as it provides insights into long-term benefit, which is a major factor in therapy selection. We discuss the clinical considerations for choosing between these therapies in the front-line setting and beyond, specifically for patients with BRAFV600 mutant melanoma based on currently available evidence. We have previously proposed a time-dependent resistance paradigm in which future therapeutic development strategies can be rooted. We also discuss how these Food and Drug Administration (FDA)-approved therapeutic modalities are being pursued earlier in the course of disease management, namely in adjuvant and neoadjuvant settings. FDA-approved interlesional oncolytic virotherapy in the modern era is also briefly discussed.


Melanoma , Proto-Oncogene Proteins B-raf , Humans , Immunotherapy , Melanoma/drug therapy , Melanoma/genetics , Mitogen-Activated Protein Kinases , Mutation/genetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics
4.
Clin Radiol ; 76(2): 154.e1-154.e9, 2021 Feb.
Article En | MEDLINE | ID: mdl-32896425

Oncology has progressed into an era of personalised medicine, whereby the therapeutic regimen is tailored to the molecular profile of the patient's cancer. Determining personalised therapeutic options is achieved by using tumour genomics and proteomics to identify the specific molecular targets against which candidate drugs can interact. Several dozen targeted drugs, many for multiple cancer types are already widely in clinical use. Molecular profiling of tumours is contingent on high-quality biopsy specimens and the most common method of tissue sampling is image-guided biopsy. Thus, for radiologists performing these biopsies, the paradigm has now shifted away from obtaining specimens simply for histopathological diagnosis to acquiring larger amounts of viable tumour cells for DNA, RNA, or protein analysis. These developments have highlighted the central role now played by radiologists in the delivery of personalised cancer care. This review describes the principles of molecular profiling assays and biopsy techniques for optimising yield, and describes a scoring system to assist in patient selection for percutaneous biopsy.


Diagnostic Imaging/methods , Genomics/methods , Neoplasms/genetics , Neoplasms/pathology , Precision Medicine/methods , Biomarkers, Tumor , Humans , Image-Guided Biopsy , Neoplasms/diagnostic imaging
5.
Ann Oncol ; 30(11): 1821-1830, 2019 11 01.
Article En | MEDLINE | ID: mdl-31504139

BACKGROUND: The National Cancer Institute-Molecular Analysis for Therapy Choice (NCI-MATCH) is a national precision medicine study incorporating centralized genomic testing to direct refractory cancer patients to molecularly targeted treatment subprotocols. This treatment subprotocol was designed to screen for potential signals of efficacy of ado-trastuzumab emtansine (T-DM1) in HER2-amplified histologies other than breast and gastroesophageal tumors. METHODS: Eligible patients had HER2 amplification at a copy number (CN) >7 based on targeted next-generation sequencing (NGS) with a custom Oncomine AmpliSeq™ (ThermoFisher Scientific) panel. Patients with prior trastuzumab, pertuzumab or T-DM1 treatment were excluded. Patients received T-DM1 at 3.6 mg/kg i.v. every 3 weeks until toxicity or disease progression. Tumor assessments occurred every three cycles. The primary end point was centrally assessed objective response rate (ORR). Exploratory end points included correlating response with HER2 CN by NGS. The impact of co-occurring genomic alterations and PTEN loss by immunohistochemistry were also assessed. RESULTS: Thirty-eight patients were enrolled and 36 included in efficacy analysis. Median prior therapies in the metastatic setting was 3 (range 0-9; unknown in one patient). Median HER2 CN was 17 (range 7-139). Partial responses were observed in two (5.6%) patients: one mucoepidermoid carcinoma of parotid gland and one parotid gland squamous cell cancer. Seventeen patients (47%) had stable disease including 8/10 (80%) with ovarian and uterine carcinomas, with median duration of 4.6 months. The 6-month progression-free survival rate was 23.6% [90% confidence interval 14.2% to 39.2%]. Common toxicities included fatigue, anemia, fever and thrombocytopenia with no new safety signals. There was a trend for tumor shrinkage with higher levels of gene CN as determined by the NGS assay. CONCLUSION: T-DM1 was well tolerated. While this subprotocol did not meet the primary end point for ORR in this heavily pre-treated diverse patient population, clinical activity was seen in salivary gland tumors warranting further study in this tumor type in dedicated trials.


Ado-Trastuzumab Emtansine/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/genetics , Neoplasms/drug therapy , Receptor, ErbB-2/genetics , Ado-Trastuzumab Emtansine/pharmacology , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/pharmacology , Drug Resistance, Neoplasm/genetics , Female , Gene Amplification , Humans , Middle Aged , National Cancer Institute (U.S.) , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Precision Medicine/methods , Progression-Free Survival , Receptor, ErbB-2/antagonists & inhibitors , United States/epidemiology
7.
Ann Oncol ; 28(10): 2581-2587, 2017 Oct 01.
Article En | MEDLINE | ID: mdl-28961848

BACKGROUND: The BRIM-3 trial showed improved progression-free survival (PFS) and overall survival (OS) for vemurafenib compared with dacarbazine in treatment-naive patients with BRAFV600 mutation-positive metastatic melanoma. We present final OS data from BRIM-3. PATIENTS AND METHODS: Patients were randomly assigned in a 1 : 1 ratio to receive vemurafenib (960 mg twice daily) or dacarbazine (1000 mg/m2 every 3 weeks). OS and PFS were co-primary end points. OS was assessed in the intention-to-treat population, with and without censoring of data for dacarbazine patients who crossed over to vemurafenib. RESULTS: Between 4 January 2010 and 16 December 2010, a total of 675 patients were randomized to vemurafenib (n = 337) or dacarbazine (n = 338, of whom 84 crossed over to vemurafenib). At the time of database lock (14 August 2015), median OS, censored at crossover, was significantly longer for vemurafenib than for dacarbazine {13.6 months [95% confidence interval (CI) 12.0-15.4] versus 9.7 months [95% CI 7.9-12.8; hazard ratio (HR) 0.81 [95% CI 0.67-0.98]; P = 0.03}, as was median OS without censoring at crossover [13.6 months (95% CI 12.0-15.4) versus 10.3 months (95% CI 9.1-12.8); HR 0.81 (95% CI 0.68-0.96); P = 0.01]. Kaplan-Meier estimates of OS rates for vemurafenib versus dacarbazine were 56% versus 46%, 30% versus 24%, 21% versus 19% and 17% versus 16% at 1, 2, 3 and 4 years, respectively. Overall, 173 of the 338 patients (51%) in the dacarbazine arm and 175 of the 337 (52%) of those in the vemurafenib arm received subsequent anticancer therapies, most commonly ipilimumab. Safety data were consistent with the primary analysis. CONCLUSIONS: Vemurafenib continues to be associated with improved median OS in the BRIM-3 trial after extended follow-up. OS curves converged after ≈3 years, likely as a result of crossover from dacarbazine to vemurafenib and receipt of subsequent anticancer therapies. CLINICALTRIALS.GOV: NCT01006980.


Indoles/therapeutic use , Melanoma/drug therapy , Mutation , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/drug therapy , Sulfonamides/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Dacarbazine/therapeutic use , Enzyme Inhibitors/therapeutic use , Female , Humans , Kaplan-Meier Estimate , Male , Melanoma/enzymology , Melanoma/genetics , Melanoma/mortality , Middle Aged , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/mortality , Treatment Outcome , Vemurafenib , Young Adult
8.
Ann Oncol ; 28(7): 1631-1639, 2017 Jul 01.
Article En | MEDLINE | ID: mdl-28475671

BACKGROUND: Previous analysis of COMBI-d (NCT01584648) demonstrated improved progression-free survival (PFS) and overall survival (OS) with combination dabrafenib and trametinib versus dabrafenib monotherapy in BRAF V600E/K-mutant metastatic melanoma. This study was continued to assess 3-year landmark efficacy and safety after ≥36-month follow-up for all living patients. PATIENTS AND METHODS: This double-blind, phase 3 study enrolled previously untreated patients with BRAF V600E/K-mutant unresectable stage IIIC or stage IV melanoma. Patients were randomized to receive dabrafenib (150 mg twice daily) plus trametinib (2 mg once daily) or dabrafenib plus placebo. The primary endpoint was PFS; secondary endpoints were OS, overall response, duration of response, safety, and pharmacokinetics. RESULTS: Between 4 May and 30 November 2012, a total of 423 of 947 screened patients were randomly assigned to receive dabrafenib plus trametinib (n = 211) or dabrafenib monotherapy (n = 212). At data cut-off (15 February 2016), outcomes remained superior with the combination: 3-year PFS was 22% with dabrafenib plus trametinib versus 12% with monotherapy, and 3-year OS was 44% versus 32%, respectively. Twenty-five patients receiving monotherapy crossed over to combination therapy, with continued follow-up under the monotherapy arm (per intent-to-treat principle). Of combination-arm patients alive at 3 years, 58% remained on dabrafenib plus trametinib. Three-year OS with the combination reached 62% in the most favourable subgroup (normal lactate dehydrogenase and <3 organ sites with metastasis) versus only 25% in the unfavourable subgroup (elevated lactate dehydrogenase). The dabrafenib plus trametinib safety profile was consistent with previous clinical trial observations, and no new safety signals were detected with long-term use. CONCLUSIONS: These data demonstrate that durable (≥3 years) survival is achievable with dabrafenib plus trametinib in patients with BRAF V600-mutant metastatic melanoma and support long-term first-line use of the combination in this setting.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Imidazoles/administration & dosage , Melanoma/drug therapy , Mutation , Oximes/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins B-raf/genetics , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Skin Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Disease Progression , Disease-Free Survival , Double-Blind Method , Drug Administration Schedule , Humans , Imidazoles/adverse effects , Imidazoles/pharmacokinetics , Kaplan-Meier Estimate , Melanoma/genetics , Melanoma/mortality , Melanoma/secondary , Oximes/adverse effects , Oximes/pharmacokinetics , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Pyridones/adverse effects , Pyridones/pharmacokinetics , Pyrimidinones/adverse effects , Pyrimidinones/pharmacokinetics , Risk Factors , Skin Neoplasms/genetics , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Time Factors , Treatment Outcome
9.
Clin Pharmacol Ther ; 100(6): 713-729, 2016 12.
Article En | MEDLINE | ID: mdl-27643536

Adaptive, seamless, multisponsor, multitherapy clinical trial designs executed as large scale platforms, could create superior evidence more efficiently than single-sponsor, single-drug trials. These trial PIPELINEs also could diminish barriers to trial participation, increase the representation of real-world populations, and create systematic evidence development for learning throughout a therapeutic life cycle, to continually refine its use. Comparable evidence could arise from multiarm design, shared comparator arms, and standardized endpoints-aiding sponsors in demonstrating the distinct value of their innovative medicines; facilitating providers and patients in selecting the most appropriate treatments; assisting regulators in efficacy and safety determinations; helping payers make coverage and reimbursement decisions; and spurring scientists with translational insights. Reduced trial times and costs could enable more indications, reduced development cycle times, and improved system financial sustainability. Challenges to overcome range from statistical to operational to collaborative governance and data exchange.


Clinical Trials as Topic/methods , Patient Selection , Reimbursement Mechanisms , Research Design , Clinical Trials as Topic/economics , Clinical Trials as Topic/organization & administration , Cooperative Behavior , Endpoint Determination , Humans , Time Factors , Translational Research, Biomedical/organization & administration
10.
Oncogene ; 35(10): 1225-35, 2016 Mar 10.
Article En | MEDLINE | ID: mdl-26073081

The mechanisms by which some melanoma cells adapt to Serine/threonine-protein kinase B-Raf (BRAF) inhibitor therapy are incompletely understood. In the present study, we used mass spectrometry-based phosphoproteomics to determine how BRAF inhibition remodeled the signaling network of melanoma cell lines that were BRAF mutant and PTEN null. Short-term BRAF inhibition was associated with marked changes in fibronectin-based adhesion signaling that were PTEN dependent. These effects were recapitulated through BRAF siRNA knockdown and following treatment with chemotherapeutic drugs. Increased fibronectin expression was also observed in mouse xenograft models as well as specimens from melanoma patients undergoing BRAF inhibitor treatment. Analysis of a melanoma tissue microarray showed loss of PTEN expression to predict for a lower overall survival, with a trend for even lower survival being seen when loss of fibronectin was included in the analysis. Mechanistically, the induction of fibronectin limited the responses of these PTEN-null melanoma cell lines to vemurafenib, with enhanced cytotoxicity observed following the knockdown of either fibronectin or its receptor α5ß1 integrin. This in turn abrogated the cytotoxic response to BRAF inhibition via increased AKT signaling, which prevented the induction of cell death by maintaining the expression of the pro-survival protein Mcl-1. The protection conveyed by the induction of FN expression could be overcome through combined treatment with a BRAF and PI3K inhibitor.


Fibronectins/metabolism , Melanoma/pathology , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Animals , Cell Line, Tumor , Cell Survival/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Integrin alpha5beta1/metabolism , Mice , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proteomics , Proto-Oncogene Proteins B-raf/deficiency , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
11.
Br J Dermatol ; 170(1): 36-44, 2014 Jan.
Article En | MEDLINE | ID: mdl-24443912

Malignant melanoma is rising in incidence. The treatment options have been very limited but advances in molecular biology and immunology have led to a greater understanding of the pathogenesis of the disease. Four drugs have been approved for the treatment of advanced melanoma in the past 2 years and two new classes of agents have recently been shown to lead to durable responses in a substantial minority of patients. The identification of biomarkers has helped clinicians and researchers segregate patients into molecular subgroups, which facilitates the selection of therapy. Preliminary work has begun on determining the ideal sequences of the various therapies. Investigations have been carried out on why these treatments work and what the mechanisms of resistance are to these therapies. It is hoped that combinations of therapies will emerge that lead to a high percentage of durable responses.


Melanoma/therapy , Skin Neoplasms/therapy , Antineoplastic Agents/therapeutic use , Humans , Melanoma/genetics , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Mutation/genetics , Protein Kinase Inhibitors/therapeutic use , Skin Neoplasms/genetics
12.
Ann Oncol ; 24(2): 530-537, 2013 Feb.
Article En | MEDLINE | ID: mdl-23035153

BACKGROUND: The cutaneous effects of rapidly accelerated fibrosarcoma kinase B (BRAF) inhibitors are not well understood. Squamous cell carcinoma (SCC), keratoacanthoma, and photosensitivity have been described in patients taking BRAF inhibitors. PATIENTS AND METHODS: To characterize the timing and frequency of skin lesions in patients receiving BRAF inhibitor therapy, we utilized a retrospective case review of 53 patients undergoing treatment with BRAF inhibitors for 4-92 weeks of therapy. Patients were evaluated at baseline, and then followed at 4- to 12-week intervals. Charts were retrospectively reviewed, and the morphology and timing of cutaneous events were recorded. RESULTS: Thirty-three of the 53 charts met exclusion/inclusion criteria, 15 were treated with vemurafenib, and 18 were treated with GSK 2118436/GSK 1120212. Of 33 patients treated with BRAF inhibitor, 13 developed photosensitivity (39.4%), 10 developed actinic keratoses (30.3%), 10 developed warts (30.3%), and 6 developed SCC (18.2%). CONCLUSIONS: Multiple cutaneous findings were observed in the 33 patients taking BRAF inhibitors. The previously described association with SCC and photosensitivity was observed in these patients as well. Over half of the observed SCCs were invasive in nature. Photosensitivity continues to be frequent with BRAF inhibitors. Patients taking BRAF inhibitors should have regular full body skin exams. Further studies are necessary to better elucidate the rates of these adverse cutaneous effects.


Antineoplastic Agents/adverse effects , Neoplasms/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Diseases/chemically induced , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/chemically induced , Female , Humans , Imidazoles/adverse effects , Imidazoles/therapeutic use , Indoles/adverse effects , Indoles/therapeutic use , Keratoacanthoma/chemically induced , Keratosis, Actinic/chemically induced , Male , Melanoma/drug therapy , Middle Aged , Oximes/adverse effects , Oximes/therapeutic use , Photosensitivity Disorders/chemically induced , Pyridones/adverse effects , Pyridones/therapeutic use , Pyrimidinones/adverse effects , Pyrimidinones/therapeutic use , Retrospective Studies , Sulfonamides/adverse effects , Sulfonamides/therapeutic use , Vemurafenib , Warts/chemically induced
13.
Br J Cancer ; 102(12): 1724-30, 2010 Jun 08.
Article En | MEDLINE | ID: mdl-20531415

BACKGROUND: Resistance to BRAF inhibitors is an emerging problem in the melanoma field. Strategies to prevent and overcome resistance are urgently required. METHODS: The dynamics of cell signalling, BrdU incorporation and cell-cycle entry after BRAF inhibition was measured using flow cytometry and western blot. The ability of combined BRAF/MEK inhibition to prevent the emergence of resistance was demonstrated by apoptosis and colony formation assays and in 3D organotypic cell culture. RESULTS: BRAF inhibition led to a rapid recovery of phospho-ERK (pERK) signalling. Although most of the cells remained growth arrested in the presence of drug, a minor population of cells retained their proliferative potential and escaped from BRAF inhibitor therapy. A function for the rebound pERK signalling in therapy escape was demonstrated by the ability of combined BRAF/MEK inhibition to enhance the levels of apoptosis and abrogate the onset of resistance. CONCLUSION: Combined BRAF/MEK inhibition may be one strategy to prevent the emergence of drug resistance in BRAF-V600E-mutated melanomas.


Drug Resistance, Neoplasm , Extracellular Signal-Regulated MAP Kinases/metabolism , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Apoptosis/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Indoles/pharmacology , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Mutation , Signal Transduction/drug effects , Sulfonamides/pharmacology , Time Factors
14.
Br J Cancer ; 100(3): 431-5, 2009 Feb 10.
Article En | MEDLINE | ID: mdl-19156138

The discovery of BRAF mutations in melanoma has not yet translated into clinical success, suggesting that BRAF/MEK inhibitors will need to be combined with other agents. In the current review, we discuss other pathways likely to be important for melanoma progression and suggest possible drug combinations for future clinical testing.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , MAP Kinase Kinase Kinases/antagonists & inhibitors , Melanoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Humans , Mice , Protein Kinase Inhibitors/pharmacology
15.
Oncogene ; 28(1): 85-94, 2009 Jan 08.
Article En | MEDLINE | ID: mdl-18794803

Here, we identify a panel of melanoma lines with non-V600E mutations in BRAF. These G469E- and D594G-mutated melanomas were found to exhibit constitutive levels of phospho-extracellular signal-regulated kinase (pERK) and low levels of phospho-mitogen-activated protein kinase/ERK kinase (pMEK) and were resistant to MEK inhibition. Upon treatment with the CRAF inhibitor sorafenib, these lines underwent apoptosis and associated with mitochondrial depolarization and relocalization of apoptosis-inducing factor, whereas the BRAF-V600E-mutated melanomas did not. Studies have shown low-activity mutants of BRAF (G469E/D594G) instead signal through CRAF. Unlike BRAF, CRAF directly regulates apoptosis through mitochondrial localization where it binds to Bcl-2 and phosphorylates BAD. The CRAF inhibitor sorafenib was found to induce a time-dependent reduction in both BAD phosphorylation and Bcl-2 expression in the D594G/G469E lines only. Knockdown of CRAF using a lentiviral shRNA suppressed both Bcl-2 expression and induced apoptosis in the D594G melanoma line but not in a V600E-mutated line. Finally, we showed in a series of xenograft studies that sorafenib was more potent at reducing the growth of tumors with the D594G mutation than those with the V600E mutation. In summary, we have identified a group of melanomas with low-activity BRAF mutations that are reliant upon CRAF-mediated survival activity.


Apoptosis , Drug Resistance, Neoplasm/genetics , Melanoma/enzymology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Apoptosis/genetics , Benzenesulfonates/pharmacology , Cell Line, Tumor , Gene Knockdown Techniques , Glutamic Acid/genetics , Glutamic Acid/metabolism , Humans , Melanoma/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Niacinamide/analogs & derivatives , Phenylurea Compounds , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-raf/genetics , Pyridines/pharmacology , RNA, Small Interfering/genetics , Signal Transduction , Sorafenib , Valine/genetics , Valine/metabolism
16.
Ann Oncol ; 18(12): 2025-9, 2007 Dec.
Article En | MEDLINE | ID: mdl-17846021

BACKGROUND: This study determined the optimally tolerated regimen (OTR) of oral lapatinib administered in combination with infusional 5-fluorouracil (5-FU), leucovorin and irinotecan (FOLFIRI) and assessed the safety, tolerability and pharmacokinetics of the combination. PATIENTS AND METHODS: Twenty-five patients were enrolled; 12 patients were treated at three dose levels to determine OTR; then 13 patients were treated at OTR to evaluate the pharmacokinetics of the combination. RESULTS: The 2-weekly OTR comprised lapatinib 1250 mg/day with irinotecan 108 mg/m(2) (day 1) and leucovorin 200 mg/m(2), 5-FU bolus 240 mg/m(2) and 5-FU infusion 360 mg/m(2) (days 1 and 2); doses of 5-FU and irinotecan represent a 40% reduction in dose compared to conventional FOLFIRI. Dose-limiting toxicities were grade 3 diarrhoea and grade 4 neutropenia. Co-administration of lapatinib increased the area under the plasma concentration-time curve of SN-38, the active metabolite of irinotecan, by an average of 41%; no other pharmacokinetic interactions were observed. Of 19 patients evaluable for disease response assessment, four patients had partial response and nine patients had stable disease. CONCLUSION: The combination of lapatinib and FOLFIRI is safe and demonstrates clinical activity; the documented PK interaction can effectively be compensated by lowering the doses of 5-FU and irinotecan. This regime may be further tested in a phase II trial.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Quinazolines/pharmacokinetics , Quinazolines/therapeutic use , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Female , Fluorouracil/administration & dosage , Humans , Irinotecan , Lapatinib , Leucovorin/administration & dosage , Male , Middle Aged , Neoplasms/drug therapy , Quinazolines/administration & dosage , Quinazolines/blood , Tandem Mass Spectrometry
17.
Br J Cancer ; 96(3): 445-9, 2007 Feb 12.
Article En | MEDLINE | ID: mdl-17245336

The loss of tumour phospho-extracellular responsive kinase (pERK) positivity is the major treatment biomarker for mitogen-activated protein kinase/extracellular responsive kinase (MEK) inhibitors. Here, we demonstrate that there is a poor correlation between pERK inhibition and the anti-proliferative effects of MEK inhibitors in melanoma cells. We suggest that Ki67 is a better biomarker for future clinical studies.


Biomarkers, Tumor/analysis , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/analysis , Ki-67 Antigen/analysis , Melanoma/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Butadienes/analysis , Cell Line, Tumor , Cell Proliferation , G1 Phase , Humans , Melanoma/pathology , Mutation , Nitriles/analysis , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins B-raf/genetics
18.
Br J Cancer ; 95(5): 581-6, 2006 Sep 04.
Article En | MEDLINE | ID: mdl-16880785

The effects of sorafenib--an oral multikinase inhibitor targeting the tumour and tumour vasculature--were evaluated in patients with advanced melanoma enrolled in a large multidisease Phase II randomised discontinuation trial (RDT). Enrolled patients received a 12-week run-in of sorafenib 400 mg twice daily (b.i.d.). Patients with changes in bi-dimensional tumour measurements <25% from baseline were then randomised to sorafenib or placebo for a further 12 weeks (ie to week 24). Patients with > or =25% tumour shrinkage after the run-in continued on open-label sorafenib, whereas those with > or =25% tumour growth discontinued treatment. This analysis focussed on secondary RDT end points: changes in bi-dimensional tumour measurements from baseline after 12 weeks and overall tumour responses (WHO criteria) at week 24, progression-free survival (PFS), safety and biomarkers (BRAF, KRAS and NRAS mutational status). Of 37 melanoma patients treated during the run-in phase, 34 were evaluable for response: one had > or =25% tumour shrinkage and remained on open-label sorafenib; six (16%) had <25% tumour growth and were randomised (placebo, n=3; sorafenib, n=3); and 27 had > or =25% tumour growth and discontinued. All three randomised sorafenib patients progressed by week 24; one remained on sorafenib for symptomatic relief. All three placebo patients progressed by week-24 and were re-started on sorafenib; one experienced disease re-stabilisation. Overall, the confirmed best responses for each of the 37 melanoma patients who received sorafenib were 19% stable disease (SD) (ie n=1 open-label; n=6 randomised), 62% (n=23) progressive disease (PD) and 19% (n=7) unevaluable. The overall median PFS was 11 weeks. The six randomised patients with SD had overall PFS values ranging from 16 to 34 weeks. The most common drug-related adverse events were dermatological (eg rash/desquamation, 51%; hand-foot skin reaction, 35%). There was no relationship between V600E BRAF status and disease stability. DNA was extracted from the biopsies of 17/22 patients. Six had V600E-positive tumours (n=4 had PD; n=1 had SD; n=1 unevaluable for response), and 11 had tumours containing wild-type BRAF (n=9 PD; n=1 SD; n=1 unevaluable for response). In conclusion, sorafenib is well tolerated but has little or no antitumour activity in advanced melanoma patients as a single agent at the dose evaluated (400 mg b.i.d.). Ongoing trials in advanced melanoma are evaluating sorafenib combination therapies.


Angiogenesis Inhibitors/therapeutic use , Benzenesulfonates/therapeutic use , Melanoma/drug therapy , Pyridines/therapeutic use , Adult , Aged , Aged, 80 and over , Angiogenesis Inhibitors/toxicity , Benzenesulfonates/toxicity , DNA Primers , Female , Genes, ras , Humans , Male , Melanoma/blood supply , Melanoma/genetics , Melanoma/pathology , Middle Aged , Neoplasm Staging , Niacinamide/analogs & derivatives , Phenylurea Compounds , Polymerase Chain Reaction , Proto-Oncogene Proteins B-raf/genetics , Pyridines/toxicity , Safety , Sorafenib
19.
Curr Opin Oncol ; 13(6): 499-505, 2001 Nov.
Article En | MEDLINE | ID: mdl-11673691

The authors review the early clinical experience with antisense oligodeoxynucleotides, documenting their limited toxicity profile and initial reports of efficacy. Several oncogene products, most notably bcl-2, c-raf-1, protein kinase C-alpha, and H-ras, have been evaluated as targets for therapeutic downregulation, and oligodeoxynucleotides designed to inhibit the expression of these products specifically have been studied extensively in phase I and II trials in cancer patients. Inhibition of target expression in tumor (non-Hodgkin lymphoma) and surrogate tissues has been demonstrated in several of these trials. Continuous infusion over 2 to 3 weeks appears preferable to weekly administration for toxicity and downregulation of target mRNA. The efficacy data available suggest that antisense therapy alone appears capable of limiting disease progression in some patients, but major tumor responses are uncommon. The specificity and tolerability of these oligodeoxynucleotides support the investigation of combinations of antisense oligodeoxynucleotides with cytotoxic chemotherapy, and early combination studies have yielded results of interest. Antisense oligodeoxynucleotides against bcl-2, c-raf-1, and protein kinase C-alpha continue to be the focus of ongoing trials.


Gene Expression Regulation, Neoplastic , Oligodeoxyribonucleotides, Antisense/therapeutic use , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Clinical Trials as Topic , Combined Modality Therapy , Disease Progression , Genes, bcl-2 , Humans , Isoenzymes , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/genetics , Melanoma/drug therapy , Melanoma/genetics , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Protein Kinase C , Protein Kinase C-alpha , Proto-Oncogene Proteins c-raf , RNA, Messenger , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics
20.
Chest ; 114(3): 801-7, 1998 Sep.
Article En | MEDLINE | ID: mdl-9743170

STUDY OBJECTIVES: To determine whether African-American race is independently associated with lung disease in scleroderma. DESIGN: Retrospective review. SETTING: University medical center in Baltimore. PATIENTS: One hundred one patients with diffuse cutaneous scleroderma with available serum samples. MEASUREMENTS: Patients underwent lung function testing as part of their routine clinical care. Percent predicted values adjusted for race were calculated for FVC, single-breath carbon monoxide diffusing capacity (Dco), and FEV1. Serum samples were assayed for the presence of antibodies to topoisomerase I and RNA polymerase II. RESULTS: Scleroderma patients of African-American race had lower percent predicted values than white patients for FVC (p<0.002), Dco (p<0.0001), and FEV1 (p<0.0001). Antibodies to topoisomerase I but not antibodies to RNA polymerase II were also associated with lung function. African-American scleroderma patients were distinct from white patients in having younger age of onset and higher prevalence of antibodies to topoisomerase I. In multivariate analyses accounting for sex, age, smoking history, years of scleroderma symptoms, and RNA polymerase II antibody status, African-American race and topoisomerase I antibody status independently predicted lower lung function. CONCLUSION: African-American race and antibodies to topoisomerase I are independent risk factors for scleroderma lung disease.


Autoantibodies/analysis , Black People , DNA Topoisomerases, Type I/immunology , Lung Diseases/immunology , Scleroderma, Systemic/immunology , Female , Forced Expiratory Volume , Humans , Lung Diseases/ethnology , Lung Diseases/physiopathology , Male , Middle Aged , Pulmonary Diffusing Capacity , RNA Polymerase II/immunology , Retrospective Studies , Risk Factors , Scleroderma, Systemic/ethnology , Scleroderma, Systemic/physiopathology , Vital Capacity
...