Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Med ; 3(12): 860-882.e15, 2022 Dec 09.
Article in English | MEDLINE | ID: mdl-36257298

ABSTRACT

BACKGROUND: The near impermeability of the blood-brain barrier (BBB) and the unique neuroimmune environment of the CNS prevents the effective use of antibodies in neurological diseases. Delivery of biotherapeutics to the brain can be enabled through receptor-mediated transcytosis via proteins such as the transferrin receptor, although limitations such as the ability to use Fc-mediated effector function to clear pathogenic targets can introduce safety liabilities. Hence, novel delivery approaches with alternative clearance mechanisms are warranted. METHODS: Binders that optimized transport across the BBB, known as transcytosis-enabling modules (TEMs), were identified using a combination of antibody discovery techniques and pharmacokinetic analyses. Functional activity of TEMs were subsequently evaluated by imaging for the ability of myeloid cells to phagocytose target proteins and cells. FINDINGS: We demonstrated significantly enhanced brain exposure of therapeutic antibodies using optimal transferrin receptor or CD98 TEMs. We found that these modules also mediated efficient clearance of tau aggregates and HER2+ tumor cells via a non-classical phagocytosis mechanism through direct engagement of myeloid cells. This mode of clearance potentially avoids the known drawbacks of FcγR-mediated antibody mechanisms in the brain such as the neurotoxic release of proinflammatory cytokines and immune cell exhaustion. CONCLUSIONS: Our study reports a new brain delivery platform that harnesses receptor-mediated transcytosis to maximize brain uptake and uses a non-classical phagocytosis mechanism to efficiently clear pathologic proteins and cells. We believe these findings will transform therapeutic approaches to treat CNS diseases. FUNDING: This research was funded by Janssen, Pharmaceutical Companies of Johnson & Johnson.


Subject(s)
Blood-Brain Barrier , Transcytosis , Blood-Brain Barrier/metabolism , Transcytosis/physiology , Receptors, Transferrin , Biological Transport/physiology , Antibodies
3.
Lancet Oncol ; 18(1): 143-154, 2017 01.
Article in English | MEDLINE | ID: mdl-27979599

ABSTRACT

BACKGROUND: We applied mathematical models to clinical trial data available at Project Data Sphere LLC (Cary, NC, USA), a non-profit universal access data-sharing warehouse. Our aim was to assess the rates of cancer growth and regression using the comparator groups of eight randomised clinical trials that enrolled patients with metastatic castration-resistant prostate cancer. METHODS: In this retrospective analysis, we used data from eight randomised clinical trials with metastatic castration-resistant prostate cancer to estimate the growth (g) and regression (d) rates of disease burden over time. Rates were obtained by applying mathematical models to prostate-specific antigen levels as the representation of tumour quantity. Rates were compared between study interventions (prednisone, mitoxantrone, and docetaxel) and off-treatment data when on-study treatment had been discontinued to understand disease behaviour during treatment and after discontinuation. Growth (g) was examined for association with a traditional endpoint (overall survival) and for its potential use as an endpoint to reduce sample size in clinical trials. FINDINGS: Estimates for g, d, or both were obtained in 2353 (88%) of 2678 patients with data available for analysis; g differentiated docetaxel (a US Food and Drug Administration-approved therapy) from prednisone and mitoxantrone and was predictive of overall survival in a landmark analysis at 8 months. A simulated sample size analysis, in which g was used as the endpoint, compared docetaxel data with mitoxantrone data and showed that small sample sizes were sufficient to achieve 80% power (16, 47, and 25 patients, respectively, in the three docetaxel comparator groups). Similar results were found when the mitoxantrone data were compared with the prednisone data (41, 39, and 41 patients in the three mitoxantrone comparator groups). Finally, after discontinuation of docetaxel therapy, median tumour growth (g) increased by nearly five times. INTERPRETATION: The application of mathematical models to existing clinical data allowed estimation of rates of growth and regression that provided new insights in metastatic castration-resistant prostate cancer. The availability of clinical data through initiatives such as Project Data Sphere, when combined with innovative modelling techniques, could greatly enhance our understanding of how cancer responds to treatment, and accelerate the productivity of clinical development programmes. FUNDING: None.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prostatic Neoplasms, Castration-Resistant/pathology , Biomarkers, Tumor/blood , Case-Control Studies , Clinical Trials, Phase III as Topic , Docetaxel , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Male , Mitoxantrone/administration & dosage , Neoplasm Staging , Prednisone/administration & dosage , Prognosis , Prostate-Specific Antigen/blood , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/drug therapy , Randomized Controlled Trials as Topic , Retrospective Studies , Survival Rate , Taxoids/administration & dosage
4.
Cancer Prev Res (Phila) ; 9(8): 635-7, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27138792

ABSTRACT

Malignant diseases develop slowly over time and are often preceded by identifiable premalignancies. As malignancy progresses, so does genomic complexity and the ability of cancers to evade most therapeutic interventions. Accordingly, with some notable exceptions, a relatively low percentage of advanced cancers are effectively treated and even fewer are cured. Despite this appreciation, much less attention has been paid to intercepting the disease process compared with that of treating well-established and refractory disease. One frequently cited reason is that the pharmaceutical industry is not interested in these pursuits. In this commentary, we attempt to define the true hurdles, the degree of difficulty inherent in each, and some important approaches to be considered. Cancer Prev Res; 9(8); 635-7. ©2016 AACR.


Subject(s)
Neoplasms/prevention & control , Primary Prevention , Drug Design , Drug Industry/economics , Humans , Noncommunicable Diseases/prevention & control , Primary Prevention/economics , Primary Prevention/trends
5.
Clin Cancer Res ; 20(17): 4449-58, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24958809

ABSTRACT

PURPOSE: Dabrafenib is a selective, potent ATP-competitive inhibitor of the BRAFV600-mutant kinase that has demonstrated efficacy in clinical trials. We report the rationale for dose selection in the first-in-human study of dabrafenib, including pharmacokinetics, tissue pharmacodynamics, 2[18F]fluoro-2-deoxy-D-glucose-positron emission tomography (FDG-PET) pharmacodynamics, and dose-response relationship. EXPERIMENTAL DESIGN: Dabrafenib was administered orally once, twice (BID), or three times daily (TID). Selected dose cohorts were expanded to collect adequate data on safety, pharmacokinetics, or pharmacodynamics. A recommended phase II dose (RP2D) was chosen based on safety, pharmacokinetic, pharmacodynamic, and response data. RESULTS: One hundred and eighty-four patients were enrolled and treated with doses ranging from 12 mg once daily to 300 mg BID in 10 cohorts. Pharmacokinetic assessment of dabrafenib demonstrated a less-than-dose-proportional increase in exposure after repeat dosing above 150 mg BID. Similar to parent drug concentrations, exposure for all metabolites demonstrated less-than-dose-proportional increases. Predicted target inhibition of pERK (>80%) was achieved at 150 mg BID, with a similar magnitude of inhibition at higher doses in BRAFV600 mutation melanoma biopsy samples. Although there was large variability between patients, FDG uptake decreased with higher daily doses in patients with BRAFV600 mutation-positive melanoma. A favorable activity and tolerability profile was demonstrated at 150 mg BID. There was no improvement with TID dosing compared with BID dosing, based on FDG-PET and tumor response analyses in patients with melanoma. CONCLUSION: The RP2D of dabrafenib was determined to be 150 mg BID after considering multiple factors, including pharmacokinetics, tissue pharmacodynamics, FDG-PET pharmacodynamics, and the dose-response relationship. A maximum tolerated dose for dabrafenib was not determined.


Subject(s)
Imidazoles/administration & dosage , Melanoma/drug therapy , Oximes/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins B-raf/genetics , Adult , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Imidazoles/adverse effects , Male , Melanoma/genetics , Melanoma/pathology , Middle Aged , Mutation , Oximes/adverse effects , Positron-Emission Tomography , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Treatment Outcome
6.
Clin Cancer Res ; 19(17): 4868-78, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23833299

ABSTRACT

PURPOSE: Dabrafenib is a selective inhibitor of V600-mutant BRAF kinase, which recently showed improved progression-free survival (PFS) as compared with dacarbazine, in metastatic melanoma patients. This study examined potential genetic markers associated with response and PFS in the phase I study of dabrafenib. EXPERIMENTAL DESIGN: Baseline (pretreatment or archival) melanoma samples were evaluated in 41 patients using a custom genotyping melanoma-specific assay, sequencing of PTEN, and copy number analysis using multiplex ligation amplification and array-based comparative genomic hybridization. Nine patients had on-treatment and/or progression samples available. RESULTS: All baseline patient samples had BRAF(V600E/K) confirmed. Baseline PTEN loss/mutation was not associated with best overall response to dabrafenib, but it showed a trend for shorter median PFS [18.3 (95% confidence interval, CI, 9.1-24.3) vs. 32.1 weeks (95% CI, 24.1-33), P=0.059]. Higher copy number of CCND1 (P=0.009) and lower copy number of CDKN2A (P=0.012) at baseline were significantly associated with decreased PFS. Although no melanomas had high-level amplification of BRAF, the two patients with progressive disease as their best response had BRAF copy gain in their tumors. CONCLUSIONS: Copy number changes in CDKN2A, CCND1, and mutation/copy number changes in PTEN correlated with the duration of PFS in patients treated with dabrafenib. The results suggest that these markers should be considered in the design and interpretation of future trials with selective BRAF inhibitors in advanced melanoma patients.


Subject(s)
Comparative Genomic Hybridization , DNA Copy Number Variations/genetics , Imidazoles/administration & dosage , Melanoma/genetics , Oximes/administration & dosage , Proto-Oncogene Proteins B-raf/genetics , Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease Progression , Disease-Free Survival , Humans , Melanoma/drug therapy , Melanoma/pathology , Mutation , Neoplasm Staging , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors
7.
Eur J Cancer ; 49(2): 395-402, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22981500

ABSTRACT

BACKGROUND: Little is known about the prevalence and clinical significance of heterogeneity of positron emission tomography with (18)F-labelled fluorodeoxyglucose-positron emission tomography (FDG-PET) response. We aim to determine the prevalence, and clinicopathologic correlates of intra-patient heterogeneity of FDG-PET response in metastatic melanoma treated with dabrafenib, and to determine whether heterogeneity predicts clinical outcome. METHODS: Patients with BRAF mutant metastatic melanoma and ≥ 2 FDG avid lesions treated on the Phase I trial of dabrafenib at a single institution (n=23) were included. FDG-PET response was assessed by comparing baseline PET scans with scans at day 15. A heterogeneous response was defined as responding and new or metabolically progressing lesion(s) in a patient, or >10% of lesions with a stable metabolic response and responding lesions in a patient. RESULTS: Six (26%) patients had a heterogeneous PET response. The median time to progression (TTP) was 7.4 months (95% confidence interval (CI): 6.5-8.3) for PET homogeneous responders and 3.0 months (95%CI: 0.6-5.4) for PET heterogeneous responders. There were no homogeneous non-responders. Age, BRAF mutation genotype, dose, and lactate dehydrogenase, did not predict for heterogeneity of PET response. Heterogeneity did not correlate with tumour response. Lung metastases were more likely to respond than other visceral metastatic sites. CONCLUSIONS: Heterogeneous FDG-PET responses are common in metastatic melanoma treated with dabrafenib, and heterogeneity is associated with a shorter TTP. FDG-PET heterogeneity may predict molecular heterogeneity, and FDG-PET directed biopsies may facilitate investigation into mechanisms of resistance to signal pathway inhibitors.


Subject(s)
Fluorodeoxyglucose F18 , Imidazoles/administration & dosage , Melanoma/diagnostic imaging , Melanoma/drug therapy , Oximes/administration & dosage , Proto-Oncogene Proteins B-raf/genetics , Radiopharmaceuticals , Adult , Dose-Response Relationship, Drug , Female , Humans , Imidazoles/adverse effects , Male , Melanoma/genetics , Melanoma/pathology , Mutation , Oximes/adverse effects , Positron-Emission Tomography/methods , Prognosis , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Survival Analysis , Treatment Outcome
8.
N Engl J Med ; 367(18): 1694-703, 2012 11 01.
Article in English | MEDLINE | ID: mdl-23020132

ABSTRACT

BACKGROUND: Resistance to therapy with BRAF kinase inhibitors is associated with reactivation of the mitogen-activated protein kinase (MAPK) pathway. To address this problem, we conducted a phase 1 and 2 trial of combined treatment with dabrafenib, a selective BRAF inhibitor, and trametinib, a selective MAPK kinase (MEK) inhibitor. METHODS: In this open-label study involving 247 patients with metastatic melanoma and BRAF V600 mutations, we evaluated the pharmacokinetic activity and safety of oral dabrafenib (75 or 150 mg twice daily) and trametinib (1, 1.5, or 2 mg daily) in 85 patients and then randomly assigned 162 patients to receive combination therapy with dabrafenib (150 mg) plus trametinib (1 or 2 mg) or dabrafenib monotherapy. The primary end points were the incidence of cutaneous squamous-cell carcinoma, survival free of melanoma progression, and response. Secondary end points were overall survival and pharmacokinetic activity. RESULTS: Dose-limiting toxic effects were infrequently observed in patients receiving combination therapy with 150 mg of dabrafenib and 2 mg of trametinib (combination 150/2). Cutaneous squamous-cell carcinoma was seen in 7% of patients receiving combination 150/2 and in 19% receiving monotherapy (P=0.09), whereas pyrexia was more common in the combination 150/2 group than in the monotherapy group (71% vs. 26%). Median progression-free survival in the combination 150/2 group was 9.4 months, as compared with 5.8 months in the monotherapy group (hazard ratio for progression or death, 0.39; 95% confidence interval, 0.25 to 0.62; P<0.001). The rate of complete or partial response with combination 150/2 therapy was 76%, as compared with 54% with monotherapy (P=0.03). CONCLUSIONS: Dabrafenib and trametinib were safely combined at full monotherapy doses. The rate of pyrexia was increased with combination therapy, whereas the rate of proliferative skin lesions was nonsignificantly reduced. Progression-free survival was significantly improved. (Funded by GlaxoSmithKline; ClinicalTrials.gov number, NCT01072175.).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Imidazoles/therapeutic use , Melanoma/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Oximes/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Pyridones/therapeutic use , Pyrimidinones/therapeutic use , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Disease-Free Survival , Drug Therapy, Combination/adverse effects , Female , Fever/chemically induced , Humans , Imidazoles/adverse effects , Imidazoles/pharmacokinetics , MAP Kinase Signaling System/drug effects , Male , Melanoma/genetics , Melanoma/secondary , Middle Aged , Mutation , Oximes/adverse effects , Oximes/pharmacokinetics , Proto-Oncogene Proteins B-raf/genetics , Pyridones/adverse effects , Pyridones/pharmacokinetics , Pyrimidinones/adverse effects , Pyrimidinones/pharmacokinetics
9.
Lancet Oncol ; 13(8): 782-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22805292

ABSTRACT

BACKGROUND: MEK is a member of the MAPK signalling cascade that is commonly activated in melanoma. Direct inhibition of MEK blocks cell proliferation and induces apoptosis. We aimed to analyse safety, efficacy, and genotyping data for the oral, small-molecule MEK inhibitor trametinib in patients with melanoma. METHODS: We undertook a multicentre, phase 1 three-part study (dose escalation, cohort expansion, and pharmacodynamic assessment). The main results of this study are reported elsewhere; here we present data relating to patients with melanoma. We obtained tumour samples to assess BRAF mutational status, and available tissues underwent exploratory genotyping analysis. Disease response was measured by Response Evaluation Criteria in Solid Tumors, and adverse events were defined by common toxicity criteria. This study is registered with ClinicalTrials.gov, number NCT00687622. FINDINGS: 97 patients with melanoma were enrolled, including 81 with cutaneous or unknown primary melanoma (36 BRAF mutant, 39 BRAF wild-type, six BRAF status unknown), and 16 with uveal melanoma. The most common treatment-related adverse events were rash or dermatitis acneiform (n=80; 82%) and diarrhoea (44; 45%), most of which were grade 2 or lower. No cutaneous squamous-cell carcinomas were recorded. Of 36 patients with BRAF mutations, 30 had not received a BRAF inhibitor before; two complete responses (both confirmed) and ten partial responses (eight confirmed) were noted in this subgroup (confirmed response rate, 33%). Median progression-free survival of this subgroup was 5·7 months (95% CI 4·0-7·4). Of the six patients who had received previous BRAF inhibition, one unconfirmed partial response was recorded. Of 39 patients with BRAF wild-type melanoma, four partial responses were confirmed (confirmed response rate, 10%). INTERPRETATION: Our data show substantial clinical activity of trametinib in melanoma and suggest that MEK is a valid therapeutic target. Differences in response rates according to mutations indicate the importance of mutational analyses in the future. FUNDING: GlaxoSmithKline.


Subject(s)
Antineoplastic Agents/administration & dosage , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Melanoma/drug therapy , Protein Kinase Inhibitors/administration & dosage , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Skin Neoplasms/drug therapy , Uveal Neoplasms/drug therapy , Administration, Oral , Adult , Aged , Antineoplastic Agents/adverse effects , DNA Mutational Analysis , Disease-Free Survival , Drug Administration Schedule , Female , Humans , Kaplan-Meier Estimate , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Male , Melanoma/enzymology , Melanoma/genetics , Melanoma/mortality , Melanoma/pathology , Middle Aged , Molecular Targeted Therapy , Mutation , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins B-raf/genetics , Pyridones/adverse effects , Pyrimidinones/adverse effects , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Time Factors , Treatment Outcome , United States , Uveal Neoplasms/enzymology , Uveal Neoplasms/genetics , Uveal Neoplasms/mortality , Uveal Neoplasms/pathology , Young Adult
10.
Lancet ; 379(9829): 1893-901, 2012 May 19.
Article in English | MEDLINE | ID: mdl-22608338

ABSTRACT

BACKGROUND: Dabrafenib is an inhibitor of BRAF kinase that is selective for mutant BRAF. We aimed to assess its safety and tolerability and to establish a recommended phase 2 dose in patients with incurable solid tumours, especially those with melanoma and untreated, asymptomatic brain metastases. METHODS: We undertook a phase 1 trial between May 27, 2009, and March 20, 2012, at eight study centres in Australia and the USA. Eligible patients had incurable solid tumours, were 18 years or older, and had adequate organ function. BRAF mutations were mandatory for inclusion later in the study because of an absence of activity in patients with wild-type BRAF. We used an accelerated dose titration method, with the first dose cohort receiving 12 mg dabrafenib daily in a 21-day cycle. Once doses had been established, we expanded the cohorts to include up to 20 patients. On the basis of initial data, we chose a recommended phase 2 dose. Efficacy at the recommended phase 2 dose was studied in patients with BRAF-mutant tumours, including those with non-Val600Glu mutations, in three cohorts: metastatic melanoma, melanoma with untreated brain metastases, and non-melanoma solid tumours. This study is registered with ClinicalTrials.gov, number NCT00880321. FINDINGS: We enrolled 184 patients, of whom 156 had metastatic melanoma. The most common treatment-related adverse events of grade 2 or worse were cutaneous squamous-cell carcinoma (20 patients, 11%), fatigue (14, 8%), and pyrexia (11, 6%). Dose reductions were necessary in 13 (7%) patients. No deaths or discontinuations resulted from adverse events, and 140 (76%) patients had no treatment-related adverse events worse than grade 2. Doses were increased to 300 mg twice daily, with no maximum tolerated dose recorded. On the basis of safety, pharmacokinetic, and response data, we selected a recommended phase 2 dose of 150 mg twice daily. At the recommended phase 2 dose in 36 patients with Val600 BRAF-mutant melanoma, responses were reported in 25 (69%, 95% CI 51·9-83·7) and confirmed responses in 18 (50%, 32·9-67·1). 21 (78%, 57·7-91·4) of 27 patients with Val600Glu BRAF-mutant melanoma responded and 15 (56%, 35·3-74·5) had a confirmed response. In Val600 BRAF-mutant melanoma, responses were durable, with 17 patients (47%) on treatment for more than 6 months. Responses were recorded in patients with non-Val600Glu BRAF mutations. In patients with melanoma and untreated brain metastases, nine of ten patients had reductions in size of brain lesions. In 28 patients with BRAF-mutant non-melanoma solid tumours, apparent antitumour activity was noted in a gastrointestinal stromal tumour, papillary thyroid cancers, non-small-cell lung cancer, ovarian cancer, and colorectal cancer. INTERPRETATION: Dabrafenib is safe in patients with solid tumours, and an active inhibitor of Val600-mutant BRAF with responses noted in patients with melanoma, brain metastases, and other solid tumours. FUNDING: GlaxoSmithKline.


Subject(s)
Antineoplastic Agents/administration & dosage , Imidazoles/administration & dosage , Neoplasms/drug therapy , Oximes/administration & dosage , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Adult , Antineoplastic Agents/adverse effects , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Carcinoma, Squamous Cell/chemically induced , Drug Administration Schedule , Fatigue/chemically induced , Female , Fever/chemically induced , Genotype , Humans , Imidazoles/adverse effects , Indoles/administration & dosage , Indoles/adverse effects , Male , Maximum Tolerated Dose , Melanoma/drug therapy , Melanoma/genetics , Melanoma/secondary , Middle Aged , Mutation/genetics , Neoplasms/genetics , Oximes/adverse effects , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/chemically induced , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Treatment Outcome , Vemurafenib
11.
Invest New Drugs ; 30(2): 662-71, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21188466

ABSTRACT

Two studies were conducted in subjects with mild or moderate hepatic or renal impairment and subjects with normal organ function to evaluate the pharmacokinetics of casopitant and to assess its safety in these populations. A total of 26 subjects were enrolled in the hepatic impairment study and 18 subjects in the renal impairment study. All subjects received oral casopitant 100 mg once-daily for 5 days. Casopitant area under the concentration-time curve (AUC) increased 11% and 24% in subjects with mild or moderate hepatic impairment, respectively, on Day 1, compared with subjects with normal hepatic function; a similar increase was observed on Day 5. The AUC of the active major metabolite, GSK525060, was reduced 29% and 19% on Days 1 and 5, respectively, in subjects with moderate hepatic impairment, but not altered by mild hepatic impairment. Casopitant AUC increased 34% and 22% on Day 1 in subjects with mild or moderate renal impairment, respectively, and 28% and 11% on Day 5, respectively, compared with subjects with normal renal function. GSK525060 AUC was increased 17% and 24% on Days 1 and 5, respectively, in subjects with mild renal impairment; but did not significantly change in subjects with moderate renal impairment. Further age-adjusted analysis showed no meaningful effect of renal impairment on casopitant or GSK525060 AUC. Plasma protein binding of casopitant and GSK525060 was similar in all subjects. The pharmacokinetics of casopitant is not altered to a clinically significant extent in subjects with mild or moderate, hepatic or renal impairment. The impact of severe hepatic or renal impairment was not evaluated.


Subject(s)
Antiemetics/pharmacokinetics , Kidney Diseases/metabolism , Liver Diseases/metabolism , Neurokinin-1 Receptor Antagonists , Piperazines/pharmacokinetics , Piperidines/pharmacokinetics , Administration, Oral , Adult , Analysis of Variance , Antiemetics/administration & dosage , Antiemetics/adverse effects , Area Under Curve , Biotransformation , Drug Administration Schedule , Female , Humans , Kidney Diseases/blood , Least-Squares Analysis , Liver Diseases/blood , Male , Middle Aged , Models, Biological , Piperazines/administration & dosage , Piperazines/adverse effects , Piperidines/administration & dosage , Piperidines/adverse effects , Protein Binding , Severity of Illness Index , United States
12.
J Transl Med ; 9: 119, 2011 Jul 25.
Article in English | MEDLINE | ID: mdl-21781349

ABSTRACT

BACKGROUND: Globally, gastric cancer is the second most common cause of cancer-related death, with the majority of the health burden borne by economically less-developed countries. METHODS: Here, we report a genetic characterization of 50 gastric adenocarcinoma samples, using affymetrix SNP arrays and Illumina mRNA expression arrays as well as Illumina sequencing of the coding regions of 384 genes belonging to various pathways known to be altered in other cancers. RESULTS: Genetic alterations were observed in the WNT, Hedgehog, cell cycle, DNA damage and epithelial-to-mesenchymal-transition pathways. CONCLUSIONS: The data suggests targeted therapies approved or in clinical development for gastric carcinoma would be of benefit to ~22% of the patients studied. In addition, the novel mutations detected here, are likely to influence clinical response and suggest new targets for drug discovery.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Mutation/genetics , Precision Medicine , Stomach Neoplasms/genetics , Cell Cycle Proteins/genetics , DNA Copy Number Variations/genetics , DNA, Neoplasm/genetics , Epithelium/metabolism , Epithelium/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genotype , Humans , Molecular Targeted Therapy , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phenotype , RNA, Neoplasm/genetics , Reproducibility of Results , Sequence Analysis, DNA , Signal Transduction/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/enzymology , Wnt Proteins/genetics , Wnt Proteins/metabolism
13.
J Clin Pharmacol ; 50(8): 951-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20124517

ABSTRACT

Casopitant, an antiemetic, is a neurokinin-1 receptor antagonist metabolized primarily by cytochrome P450 3A4 (CYP3A4). Three phase 1 studies with 131 healthy subjects examined the impact of a strong CYP3A inhibitor (ketoconazole) and inducer (rifampin) on the pharmacokinetics and safety of casopitant. Oral casopitant was administered alone (study 1, 100-mg single dose; study 2, 150 mg on day 1, 50 mg on days 2 and 3; study 3, 150-mg single dose) with either 400 mg daily of oral ketoconazole or 600 mg daily of oral rifampin. Ketoconazole increased the maximum observed plasma concentration (C(max)) and area under the plasma concentration time curve to the last sampling time, t (AUC(0-t)) of single-dose casopitant 2.7-fold and 12-fold and increased the C(max) of 3-day casopitant 2.5-fold on day 1 and 2.9-fold on day 3, whereas AUC((0-tau)) increased 4.3-fold on day 1 and 5.8-fold on day 3. Neither safety signals nor prolongation of Fredericia-corrected QT was observed at these increased exposures in study 2. Repeat-dose rifampin reduced the C(max) and AUC((0-t)) of casopitant 96% and 90%, respectively. These clinical studies confirmed the role of CYP3A in the metabolism and disposition of casopitant. Coadministration of casopitant with strong inhibitors of CYP3A is likely to increase plasma exposure of casopitant, whereas coadministration with strong inducers of CYP3A is likely to decrease casopitant exposure and compromise efficacy.


Subject(s)
Antiemetics/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors , Ketoconazole/pharmacology , Neurokinin-1 Receptor Antagonists , Piperazines/pharmacokinetics , Piperidines/pharmacokinetics , Rifampin/pharmacology , Adolescent , Adult , Antiemetics/adverse effects , Area Under Curve , Electrocardiography , Enzyme Induction , Humans , Middle Aged , Piperazines/adverse effects , Piperidines/adverse effects , Young Adult
14.
J Clin Pharmacol ; 50(3): 268-75, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19808951

ABSTRACT

Topotecan pharmacokinetics at higher infusion rates (4 mg/m2 over 30 minutes) have not been studied. The authors report a pharmacokinetics and safety study of this dose in advanced cancer patients. Sixteen patients were given a 4-mg/m2 topotecan infusion intravenously (IV) over 30 minutes weekly for 3 weeks, repeated every 28 days. Pharmacokinetics were determined after the first dose. Plasma concentrations of total topotecan were measured to derive CL, V(ss), C(max), t(max), t(1/2), AUC(0-t), and AUC(0-infinity). Plasma total topotecan concentrations decreased biexponentially, with a mean CL value of 20.6 L/h, V(ss) value of 101 L, and t(1/2) value of 5.0 h. Nine significant adverse events (all hematologic) were topotecan related. Grade 3 or less adverse events included anemia, thrombocytopenia, leukopenia, and fatigue. Pharmacokinetics of the 4-mg/m2 infusion of topotecan over 30 minutes are comparable to findings from studies of lower and higher doses. Toxicities are similar to previous reports.


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Drug Tolerance , Neoplasms/drug therapy , Topotecan/adverse effects , Topotecan/pharmacokinetics , Adult , Aged , Aged, 80 and over , Anemia/chemically induced , Antineoplastic Agents/administration & dosage , Biological Availability , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Drug Administration Schedule , Fatigue/chemically induced , Female , Humans , Infusions, Intravenous , Leukopenia/chemically induced , Male , Metabolic Clearance Rate , Middle Aged , Thrombocytopenia/chemically induced , Topotecan/administration & dosage
15.
Breast Cancer Res Treat ; 119(3): 685-99, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20012355

ABSTRACT

Neoadjuvant chemotherapy has been shown to be equivalent to post-operative treatment for breast cancer, and allows for assessment of chemotherapy response. In a pilot trial of docetaxel (T) and capecitabine (X) neoadjuvant chemotherapy for Stage II/III BC, we assessed correlation between baseline gene expression and tumor response to treatment, and examined changes in gene expression associated with treatment. Patients received four cycles of TX. Tumor tissue obtained from Mammotome core biopsies pretreatment (BL) and post-cycle 1 (C1) of TX was FLash frozen and stored at -70 degrees C until processing. Gene expression analysis utilized Affymetrix HG-U133 Plus 2.0 GeneChip arrays. Statistical analysis was performed using BRB Array Tools after RMA normalization. Gene ontology (GO) pathway analysis used random variance t tests with a significance level of P\0.005. For gene categories identified byGO pathway analysis as significant, expression levels of individual genes within those pathways were compared between classes using univariate t tests; those genes with significance level of P\0.05 were reported. PAM50 analyses were performed on tumor samples to investigate biologic subtype and risk of relapse (ROR). Using GO pathway analysis, 39 gene categories discriminated between responders and non-responders,most notably genes involved in microtubule assembly and regulation. When comparing pre- and post-chemotherapy specimens, we identified 71 differentially expressed gene categories, including DNA repair and cell proliferation regulation. There were 45 GO pathways in which the change in expression after one cycle of chemotherapy was significantly different among responders and nonresponders. The majority of tumor samples fell into the basal like and luminal B categories. ROR scores decreased in response to chemotherapy; this change was more evident in samples from patients classified as responders by clinical criteria. GO pathway analysis identified a number of gene categories pertinent to therapeutic response, and may be an informative method for identifying genes important in response to chemotherapy. Larger studies using the methods described here are necessary to fully evaluate gene expression changes in response to chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Breast Neoplasms/pathology , Capecitabine , Cluster Analysis , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Docetaxel , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Gene Expression , Humans , Neoadjuvant Therapy , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Pilot Projects , Taxoids/administration & dosage
16.
Support Care Cancer ; 17(9): 1177-85, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19205755

ABSTRACT

INTRODUCTION: Pharmacokinetic interactions between casopitant (a substrate and weak to moderate inhibitor of CYP3A), dexamethasone (a substrate and weak inducer of CYP3A), and ondansetron (a mixed CYP substrate) were evaluated in a two-part, three-period, single-sequence study in two groups of healthy subjects. MATERIALS AND METHODS: Part 1: subjects received oral casopitant (regimen A); oral dexamethasone and IV ondansetron (regimen B); and oral casopitant, a reduced dose of oral dexamethasone, and IV ondansetron (regimen C). Part 2: subjects received oral casopitant (regimen D); IV dexamethasone and oral ondansetron (regimen E); and oral casopitant, IV dexamethasone, and oral ondansetron (regimen F). Each regimen was separated by 14 days. RESULTS: Casopitant AUC in regimen C was increased 28% on day 1 but decreased 34% on day 3 compared to casopitant alone in regimen A. When given with casopitant and ondansetron in regimen C, dexamethasone AUC was 17% lower on day 1, but similar on day 3, compared to regimen B (representing dose-normalized increases in exposure of 39% and 108%, respectively). Ondansetron exposure was equivalent in regimens B and C. Casopitant AUC in regimen F was similar to regimen D on days 1 and 3. Dexamethasone AUC increased 21% when given with oral casopitant and oral ondansetron (regimen F compared to regimen E). Ondansetron exposure was equivalent in regimens E and F. CONCLUSION: When repeat-dose oral dexamethasone is to be coadministered with oral casopitant, a reduction in dexamethasone dose may be considered; however, no change in casopitant dose is required. Ondansetron exposure was not affected by coadministration with casopitant.


Subject(s)
Antiemetics/pharmacokinetics , Dexamethasone/pharmacokinetics , Ondansetron/pharmacokinetics , Piperazines/pharmacology , Piperidines/pharmacology , Adolescent , Adult , Antiemetics/administration & dosage , Antiemetics/pharmacology , Dexamethasone/administration & dosage , Drug Therapy, Combination , Drug-Related Side Effects and Adverse Reactions , Female , Humans , Male , Middle Aged , Nausea/chemically induced , Nausea/prevention & control , Neurokinin-1 Receptor Antagonists , Ondansetron/administration & dosage , Piperazines/administration & dosage , Piperidines/administration & dosage , Vomiting/chemically induced , Vomiting/prevention & control , Young Adult
17.
Support Care Cancer ; 17(9): 1187-93, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19205754

ABSTRACT

OBJECTIVE: The objective of this study was to characterize the impact of casopitant, a novel neurokinin-1 receptor antagonist under investigation for the prevention of postoperative and chemotherapy-induced nausea and vomiting, on the pharmacokinetics of the commonly prescribed 5-hydroxytryptamine receptor 3 receptor antagonists, dolasetron or granisetron. MATERIALS AND METHODS: In a phase I, open-label, two-part, two-period, single-sequence study, two cohorts of healthy subjects received either oral dolasetron (100 mg once daily for 3 days) or oral granisetron (2 mg once daily for 3 days) alone (period 1) and combined with oral casopitant, 150 mg day 1, 50 mg days 2 and 3 (period 2). Pharmacokinetics of hydrodolasetron and granisetron were assessed on days 1 and 3 of each period. Log-transformed area under the curve (AUC) and Cmax were statistically analyzed by performing an analysis of variance. Eighteen subjects were enrolled in the dolasetron cohort; nine subjects were CYP2D6 extensive metabolizers (EMs) and nine subjects were CYP2D6 poor metabolizers. Nineteen subjects were enrolled in the granisetron cohort. RESULTS: The largest changes in hydrodolasetron exposure after coadministration with casopitant were seen in CYP2D6 EMs, with a 24% increase in hydrodolasetron AUC on day 1 and 30% increase in Cmax on days 1 and 3. All other changes in hydrodolasetron exposure were <20%, and granisetron exposure was not altered to any relevant extent (<11%). CONCLUSION: None of the changes observed are considered clinically meaningful, and coadministration of casopitant with dolasetron or granisetron was well tolerated.


Subject(s)
Antiemetics/pharmacokinetics , Granisetron/pharmacokinetics , Indoles/pharmacokinetics , Piperazines/pharmacology , Piperidines/pharmacology , Quinolizines/pharmacokinetics , Adolescent , Adult , Antiemetics/administration & dosage , Antiemetics/pharmacology , Drug Therapy, Combination , Drug-Related Side Effects and Adverse Reactions , Female , Granisetron/administration & dosage , Humans , Indoles/administration & dosage , Male , Middle Aged , Nausea/chemically induced , Nausea/prevention & control , Piperazines/administration & dosage , Piperidines/administration & dosage , Quinolizines/administration & dosage , Vomiting/chemically induced , Vomiting/prevention & control , Young Adult
18.
J Clin Oncol ; 26(20): 3317-23, 2008 Jul 10.
Article in English | MEDLINE | ID: mdl-18490651

ABSTRACT

PURPOSE: The combination of lapatinib and trastuzumab has been observed to have a synergistic, antiproliferative effect against ErbB2-positive breast cancer cells in vitro. This phase I study assessed the safety, clinical feasibility, optimally tolerated regimen (OTR), pharmacokinetics (PK), and preliminary clinical activity of this combination in patients with ErbB2-positive advanced breast cancer. PATIENTS AND METHODS: Cohorts of three patients with ErbB2-positive advanced breast cancer were treated with escalating doses of lapatinib (750 to 1,500 mg) administered once daily (continuous) in combination with trastuzumab (4 mg/kg loading dose then 2 mg/kg weekly) to determine the OTR. Once the OTR was determined, additional patients were enrolled to provide the PK profile of both agents alone and in combination. RESULTS: A total of 54 patients were treated: 27 in the dose-escalation group and 27 in the PK group. Overall, adverse events were mild to moderate in severity, with no drug-related grade 4 events. The most frequent drug-related grade 3 events included diarrhea (17%), fatigue (11%), and rash (6%). The OTR was 1,000 mg lapatinib with standard weekly trastuzumab. One patient had a complete response and seven patients had partial responses. The PK parameters (maximum concentration in plasma and area under the curve) of lapatinib and trastuzumab in combination were not significantly different than when either was administered alone. CONCLUSION: The OTR of the lapatinib/trastuzumab combination was lapatinib 1,000 mg per day with standard weekly trastuzumab. At these doses, the regimen was well tolerated and clinically active in this heavily pretreated ErbB2-positive breast cancer population.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Neoplasm Invasiveness/pathology , Receptor, ErbB-2/metabolism , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Infusions, Intravenous , Lapatinib , Maximum Tolerated Dose , Middle Aged , Quinazolines/administration & dosage , Quinazolines/pharmacokinetics , Risk Assessment , Survival Analysis , Trastuzumab , Treatment Outcome
19.
Cancer ; 111(3): 173-84, 2007 Jun 25.
Article in English | MEDLINE | ID: mdl-17487852

ABSTRACT

BACKGROUND: There is an unmet clinical need for economic, minimally invasive procedures that use a limited number of cells for the molecular profiling of tumors in individual patients. Reverse-phase protein microarray (RPPM) technology has been applied successfully to the quantitative analysis of breast, ovarian, prostate, and colorectal cancers using frozen surgical specimens. METHODS: For this report, the authors investigated the novel use of RPPM technology for the analysis of both archival cytology aspirate smears and frozen fine-needle aspiration (FNA) samples. RPPMs were printed with 63 breast FNA samples that were obtained before, during, and after treatment from 21 patients who were enrolled in a Phase II trial of neoadjuvant capecitabine and docetaxel therapy for breast cancer. RESULTS: Based on an MCF7 cell line model of breast adenocarcinoma, the sensitivity of the RPPM detection method was in the femtomolar range with a coefficient of variance <13.5% for the most dilute sample. Assay linearity was noted from 1.0 microg/microL to 7.8 ng/microL total protein/array spot (R(2) = 0.9887) for a membrane receptor protein (epidermal growth factor receptor; R(2) = 0.9935). CONCLUSIONS: The results from this study indicated that low-abundance analytes and phosphorylated and nonphosphorylated proteins in specimens that consist of a few thousand cells obtained through FNA can be quantified with RPPM technology. The ability to monitor the in vivo state of cell-signaling proteins before and after treatment potentially will augment the ability to design individualized therapy regimens through the mapping of aberrant cell-signaling phenotypes. The mapping of these protein pathways will further the development of rational drug targets.


Subject(s)
Breast Neoplasms/metabolism , Microarray Analysis/methods , Proteome/analysis , Proteomics/methods , Bayes Theorem , Biopsy, Fine-Needle/methods , Breast/metabolism , Breast/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Cluster Analysis , Female , Humans , Phosphoproteins/analysis , Protein Kinases/analysis , Reproducibility of Results
20.
Int J Radiat Oncol Biol Phys ; 63(5): 1400-12, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16005577

ABSTRACT

PURPOSE: To examine the effects the proteasome inhibitor bortezomib (VELCADE) on transcription factor nuclear factor-kappaB (NF-kappaB) and target genes and the feasibility of combination therapy with reirradiation in patients with recurrent head-and-neck squamous cell carcinoma (HNSCC). METHODS AND MATERIALS: The tolerability and response to bortezomib 0.6 mg/m2 and 0.9 mg/m2 given twice weekly concurrent with daily reirradiation to 50-70 Gy was explored. Blood proteasome inhibition and NF-kappaB-modulated cytokines and factors were measured. Proteasome inhibition, nuclear localization of NF-kappaB phospho-p65, apoptosis, and expression of NF-kappaB-modulated mRNAs were compared in serial biopsies from accessible tumors. RESULTS: The maximally tolerated dose was exceeded, and study was limited to 7 and 2 patients, respectively, given bortezomib 0.6 mg/m2 and 0.9 mg/m2/dose with reirradiation. Grade 3 hypotension and hyponatremia were dose limiting. Mucositis was Grade 3 or less and was delayed. The mean blood proteasome inhibition at 1, 24, and 48 h after 0.6 mg/m2 was 32%, 16%, and 7% and after 0.9 mg/m2 was 56%, 26%, and 14%, respectively. Differences in proteasome and NF-kappaB activity, apoptosis, and expression of NF-kappaB-modulated cell cycle, apoptosis, and angiogenesis factor mRNAs were detected in 2 patients with minor tumor reductions and in serum NF-kappaB-modulated cytokines in 1 patient with a major tumor reduction. CONCLUSIONS: In combination with reirradiation, the maximally tolerated dose of bortezomib was exceeded at a dose of 0.6 mg/m2 and the threshold of proteasome inhibition. Although this regimen with reirradiation is not feasible, bortezomib induced detectable differences in NF-kappaB localization, apoptosis, and NF-kappaB-modulated genes and cytokines in tumor and serum in association with tumor reduction, indicating that other schedules of bortezomib combined with primary radiotherapy or reirradiation may merit future investigation.


Subject(s)
Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Carcinoma, Squamous Cell/therapy , Gene Expression/drug effects , Head and Neck Neoplasms/therapy , NF-kappa B/antagonists & inhibitors , Neoplasm Recurrence, Local/therapy , Protease Inhibitors/therapeutic use , Pyrazines/therapeutic use , Adult , Aged , Apoptosis/drug effects , Apoptosis/genetics , Bortezomib , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/radiotherapy , Cytokines/blood , Female , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/radiotherapy , Humans , Male , Middle Aged , Mouth Mucosa , NF-kappa B/analysis , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/radiotherapy , Stomatitis/etiology , Transcription Factor RelA/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...