Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Clin Cancer Res ; 22(6): 1371-7, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26546620

ABSTRACT

PURPOSE: Pazopanib is an effective treatment for advanced renal cell carcinoma and soft-tissue sarcoma. Transaminase elevations have been commonly observed in pazopanib-treated patients. We conducted pharmacogenetic analyses to explore mechanistic insight into pazopanib-induced liver injury. EXPERIMENTAL DESIGN: The discovery analysis tested association between four-digit HLA alleles and alanine aminotransferase (ALT) elevation in pazopanib-treated patients with cancer from eight clinical trials (N = 1,188). We conducted confirmatory analysis using an independent dataset of pazopanib-treated patients from 23 additional trials (N = 1,002). Genome-wide association study (GWAS) for transaminase elevations was also conducted. RESULTS: The discovery study identified an association between HLA-B*57:01 carriage and ALT elevation [P = 5.0 × 10(-5) for maximum on-treatment ALT (MaxALT); P = 4.8 × 10(-4) for time to ALT > 3× upper limit of normal (ULN) event; P = 4.1 × 10(-5) for time to ALT > 5× ULN event] that is significant after adjustment for number of HLA alleles tested. We confirmed these associations with time to ALT elevation event (P = 8.1 × 10(-4) for ALT > 3× ULN, P = 9.8 × 10(-3) for ALT > 5× ULN) in an independent dataset. In the combined data, HLA-B*57:01 carriage was associated with ALT elevation (P = 4.3 × 10(-5) for MaxALT, P = 5.1 × 10(-6) for time to ALT > 3×ULN event, P = 5.8 × 10(-6) for time to ALT > 5× ULN event). In HLA-B*57:01 carriers and noncarriers, frequency of ALT > 3× ULN was 31% and 19%, respectively, and frequency of ALT > 5× ULN was 18% and 10%, respectively. GWAS revealed a possible borderline association, which requires further evaluation. CONCLUSIONS: These data indicate that HLA-B*57:01 carriage confers higher risk of ALT elevation in patients receiving pazopanib and provide novel insight implicating an immune-mediated mechanism for pazopanib-associated hepatotoxicity in some patients.


Subject(s)
Alleles , Antineoplastic Agents/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Genetic Predisposition to Disease , HLA-B Antigens/genetics , Neoplasms/complications , Pyrimidines/adverse effects , Sulfonamides/adverse effects , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Chemical and Drug Induced Liver Injury/diagnosis , Chemical and Drug Induced Liver Injury/metabolism , Female , HLA-B Antigens/chemistry , Heterozygote , Humans , Indazoles , Liver Function Tests , Male , Middle Aged , Models, Molecular , Molecular Conformation , Neoplasms/drug therapy , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/therapeutic use , Young Adult
2.
Clin Cancer Res ; 21(2): 365-72, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25411163

ABSTRACT

PURPOSE: VEGF receptor (VEGFR) kinases are important drug targets in oncology that affect function of systemic endothelial cells. To discover genetic markers that affect VEGFR inhibitor pharmacodynamics, we performed a genome-wide association study of serum soluble vascular VEGFR2 concentrations [sVEGFR2], a pharmacodynamic biomarker for VEGFR2 inhibitors. EXPERIMENTAL DESIGN: We conducted a genome-wide association study (GWAS) of [sVEGFR2] in 736 healthy Old Order Amish volunteers. Gene variants identified from the GWAS were genotyped serially in a cohort of 128 patients with advanced solid tumor with baseline [sVEGFR2] measurements, and in 121 patients with renal carcinoma with [sVEGFR2] measured before and during pazopanib therapy. RESULTS: rs34231037 (C482R) in KDR, the gene encoding sVEGFR2 was found to be highly associated with [sVEGFR2], explaining 23% of the variance (P = 2.7 × 10(-37)). Association of rs34231037 with [sVEGFR2] was replicated in 128 patients with cancer with comparable effect size (P = 0.025). Furthermore, rs34231037 was a significant predictor of changes in [sVEGFR2] in response to pazopanib (P = 0.01). CONCLUSION: Our findings suggest that genome-wide analysis of phenotypes in healthy populations can expedite identification of candidate pharmacogenetic markers. Genotyping for germline variants in KDR may have clinical utility in identifying patients with cancer with unusual sensitivity to effects of VEGFR2 kinase inhibitors.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Vascular Endothelial Growth Factor Receptor-2/genetics , Adolescent , Angiogenesis Inhibitors/therapeutic use , Carcinoma, Renal Cell/blood , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Female , Genome-Wide Association Study , Humans , Indazoles , Kidney Neoplasms/blood , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Male , Polymorphism, Single Nucleotide , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Vascular Endothelial Growth Factor Receptor-2/blood , Young Adult
3.
N Engl J Med ; 369(8): 722-31, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23964934

ABSTRACT

BACKGROUND: Pazopanib and sunitinib provided a progression-free survival benefit, as compared with placebo or interferon, in previous phase 3 studies involving patients with metastatic renal-cell carcinoma. This phase 3, randomized trial compared the efficacy and safety of pazopanib and sunitinib as first-line therapy. METHODS: We randomly assigned 1110 patients with clear-cell, metastatic renal-cell carcinoma, in a 1:1 ratio, to receive a continuous dose of pazopanib (800 mg once daily; 557 patients) or sunitinib in 6-week cycles (50 mg once daily for 4 weeks, followed by 2 weeks without treatment; 553 patients). The primary end point was progression-free survival as assessed by independent review, and the study was powered to show the noninferiority of pazopanib versus sunitinib. Secondary end points included overall survival, safety, and quality of life. RESULTS: Pazopanib was noninferior to sunitinib with respect to progression-free survival (hazard ratio for progression of disease or death from any cause, 1.05; 95% confidence interval [CI], 0.90 to 1.22), meeting the predefined noninferiority margin (upper bound of the 95% confidence interval, <1.25). Overall survival was similar (hazard ratio for death with pazopanib, 0.91; 95% CI, 0.76 to 1.08). Patients treated with sunitinib, as compared with those treated with pazopanib, had a higher incidence of fatigue (63% vs. 55%), the hand-foot syndrome (50% vs. 29%), and thrombocytopenia (78% vs. 41%); patients treated with pazopanib had a higher incidence of increased levels of alanine aminotransferase (60%, vs. 43% with sunitinib). The mean change from baseline in 11 of 14 health-related quality-of-life domains, particularly those related to fatigue or soreness in the mouth, throat, hands, or feet, during the first 6 months of treatment favored pazopanib (P<0.05 for all 11 comparisons). CONCLUSIONS: Pazopanib and sunitinib have similar efficacy, but the safety and quality-of-life profiles favor pazopanib. (Funded by GlaxoSmithKline Pharmaceuticals; COMPARZ ClinicalTrials.gov number, NCT00720941.).


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Renal Cell/drug therapy , Indoles/therapeutic use , Kidney Neoplasms/drug therapy , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Sulfonamides/therapeutic use , Adult , Angiogenesis Inhibitors/adverse effects , Carcinoma, Renal Cell/secondary , Disease-Free Survival , Double-Blind Method , Female , Humans , Indazoles , Indoles/adverse effects , Kaplan-Meier Estimate , Kidney Neoplasms/pathology , Male , Pyrimidines/adverse effects , Pyrroles/adverse effects , Quality of Life , Sulfonamides/adverse effects , Sunitinib
4.
Invest New Drugs ; 31(3): 751-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23054212

ABSTRACT

This phase I, open-label, dose-escalation study assessed the maximum-tolerated dose, safety, pharmacokinetics, and preliminary antitumor activity of pazopanib plus lapatinib combination therapy in patients with solid tumors. Patients were to take pazopanib and lapatinib orally once daily in a fasting condition. During the escalation phase, pazopanib and lapatinib doses were escalated in serial patient cohorts, and a limited blood sampling scheme was applied for pharmacokinetic evaluation. In the expansion phase, potential pharmacokinetic interaction between pazopanib and lapatinib was evaluated more extensively. Seventy-five patients were treated. Multiple dosing levels were studied, combining pazopanib up to 800 mg/day with lapatinib up to 1,500 mg/day. Dose-limiting toxicities observed included grade 3 neutropenia, fatigue, asymptomatic decline in left ventricular ejection fraction, diarrhea, and liver enzyme elevations. The most common drug-related adverse events were diarrhea, nausea, anorexia, fatigue, vomiting, rash, hair depigmentation, and hypertension. The dose recommended for further evaluation was pazopanib 800 mg plus lapatinib 1,500 mg (paz-800/lap-1500). No clinically significant drug-drug interaction was observed at the paz-400/lap-1000 level. However, at paz-800/lap-1500, an increase in both the AUC0-t and Cmax of pazopanib was observed. Four partial responses were observed in patients with renal cancer (n=2), giant-cell tumor of the bone (n=1), and thyroid cancer (n=1). Stable disease for ≥ 18 weeks was seen in 12 patients. Pazopanib and lapatinib can be administered in combination at their respective single-agent doses with an acceptable safety profile. Further evaluation of the combination will be pursued, exploring both paz-800/lap-1500 and paz-400/lap-1000.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasms/drug therapy , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Female , Humans , Indazoles , Lapatinib , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/blood , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Quinazolines/administration & dosage , Quinazolines/adverse effects , Quinazolines/pharmacokinetics , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Sulfonamides/pharmacokinetics , Young Adult
5.
Lancet Oncol ; 13(8): 827-37, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22759480

ABSTRACT

BACKGROUND: Several targeted drugs are approved for treatment of patients with metastatic renal-cell cancer, but no validated biomarkers are available for prediction of clinical outcome. We aimed to assess the prognostic and predictive associations of pretreatment plasma concentrations of cytokine and angiogenic factors (CAFs) with data from a phase 2 and a phase 3 trial of pazopanib treatment. METHODS: We used a three-step approach for screening, confirmation, and validation of prospective CAF biomarkers. We screened 17 CAFs in 129 patients who had the greatest or least tumour shrinkage in a phase 2 trial of 215 patients treated with pazopanib. We confirmed associations of candidate CAFs (those identified in the screening and from previous studies) with tumour response and progression-free survival (PFS) in 215 patients from this phase 2 trial with an independent analytical platform. We validated confirmed markers in 344 patients from a randomised, placebo-controlled, phase 3 clinical study of pazopanib. FINDINGS: Five candidate markers emerged from initial screening-interleukin 6, interleukin 8, hepatocyte growth factor (HGF), tissue inhibitor of metalloproteinases (TIMP)-1, and E-selectin. Confirmatory analyses identified associations of interleukin 6, interleukin 8, VEGF, osteopontin, E-selectin, and HGF with continuous tumour shrinkage or PFS in patients treated with pazopanib. In the validation set of samples from the phase 3 trial, patients treated with pazopanib who had high concentrations (relative to median) of interleukin 8 (p=0·006), osteopontin (p=0·0004), HGF (p=0·010), and TIMP-1 (p=0·006) had shorter PFS than did those with low concentrations. In the placebo group, high concentrations of interleukin 6 (p<0·0001), interleukin 8 (p=0·002), and osteopontin (p<0·0001) were all prognostically associated with shorter PFS. These factors were stronger prognostic markers than were standard clinical classifications (Eastern Cooperative Oncology Group, Memorial Sloan-Kettering Cancer Center, and Heng criteria). High concentrations of interleukin 6 were predictive of improved relative PFS benefit from pazopanib compared with placebo (p(interaction)=0·009); standard clinical classifications were not predictive of PFS benefit. INTERPRETATION: CAF profiles could provide prognostic information beyond that of standard clinical classification and identify markers predictive of pazopanib benefit in patients with metastatic renal-cell carcinoma. Further studies of the predictive effects of these markers in different populations and with different drugs (eg, mTOR inhibitors) are warranted. FUNDING: GlaxoSmithKline.


Subject(s)
Angiogenic Proteins/blood , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/blood , Carcinoma, Renal Cell/drug therapy , Cytokines/blood , Kidney Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Carcinoma, Renal Cell/blood , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/secondary , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Cluster Analysis , Disease-Free Survival , Humans , Indazoles , Kaplan-Meier Estimate , Kidney Neoplasms/blood , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Linear Models , Logistic Models , Multicenter Studies as Topic , Predictive Value of Tests , Proportional Hazards Models , Randomized Controlled Trials as Topic , Retrospective Studies , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome , Tumor Burden
7.
J Clin Oncol ; 29(18): 2557-64, 2011 Jun 20.
Article in English | MEDLINE | ID: mdl-21576632

ABSTRACT

PURPOSE: Pazopanib, an oral angiogenesis inhibitor, is approved for the treatment of advanced renal cell carcinoma (RCC). Response to pazopanib monotherapy varies between patients, and no validated biomarkers predictive of treatment outcome have been identified. We tested the hypothesis that this variability is partially dependent on germline genetic variants that may affect pazopanib exposure or angiogenesis pathways. PATIENTS AND METHODS: Twenty-seven functional polymorphisms within 13 genes were evaluated in 397 patients with RCC. Genetic association with progression-free survival (PFS) and objective response rate (RR) was analyzed using the Cox proportional hazards model and proportional odds model, respectively. RESULTS: Three polymorphisms in IL8 and HIF1A and five polymorphisms in HIF1A, NR1I2, and VEGFA showed nominally significant association (P ≤ .05) with PFS and RR, respectively. Compared with the wild-type AA genotype (median PFS, 48 weeks), the IL8 2767TT variant genotype showed inferior PFS (27 weeks, P = .009). The HIF1A 1790AG genotype was associated with inferior PFS and reduced RR, compared with the wild-type GG genotype (median PFS, 20 v 44 weeks; P = .03; RR, 30% v 43%, P = .02). Reductions in RR were detected for the NR1I2 -25385TT genotype, compared with the wild-type CC genotype (37% v 50%, P = .03), and for the VEGFA -1498CC genotype compared with the TT genotypes (33% v 51%). CONCLUSION: Germline variants in angiogenesis- and exposure-related genes may predict treatment response to pazopanib monotherapy in patients with RCC. If validated, these markers may explain why certain patients fail antiangiogenesis therapy and they may support the use of alternative strategies to circumvent this issue.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Neoplasm Proteins/genetics , Neovascularization, Physiologic/genetics , Polymorphism, Single Nucleotide , Pyrimidines/therapeutic use , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Sulfonamides/therapeutic use , Angiogenesis Inhibitors/pharmacology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Clinical Trials, Phase II as Topic/statistics & numerical data , Cross-Over Studies , DNA Mutational Analysis , Disease-Free Survival , Genetic Markers , Genotype , Germ-Line Mutation , Humans , Indazoles , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Multicenter Studies as Topic/statistics & numerical data , Proportional Hazards Models , Pyrimidines/pharmacology , Randomized Controlled Trials as Topic/statistics & numerical data , Sulfonamides/pharmacology
8.
J Hepatol ; 54(6): 1237-43, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21145803

ABSTRACT

BACKGROUND & AIMS: Pazopanib has demonstrated clinical benefit in patients with advanced renal cell carcinoma (RCC) and is generally well tolerated. However, transaminase elevations have commonly been observed. This 2-stage study sought to identify genetic determinants of alanine transaminase (ALT) elevations in pazopanib-treated white patients with RCC. METHODS: Data from two separate clinical studies were used to examine the association of genetic polymorphisms with maximum on-treatment ALT levels. RESULTS: Of 6852 polymorphisms in 282 candidate genes examined in an exploratory dataset of 115 patients, 92 polymorphisms in 40 genes were significantly associated with ALT elevation (p<0.01). Two markers (rs2858996 and rs707889) in the HFE gene, which are not yet known to be associated with hemochromatosis, showed evidence for replication. Because of multiple comparisons, there was a 12% likelihood the replication occurred by chance. These two markers demonstrated strong linkage disequilibrium (r(2)=0.99). In the combined dataset, median (25-75th percentile) maximum ALT values were 1.2 (0.7-1.9), 1.1 (0.8-2.5), and 5.4 (1.9-7.6)×ULN for rs2858996 GG (n=148), GT (n=82), and TT (n=1 2) genotypes, respectively. All 12 TT patients had a maximum ALT>ULN, and 8 (67%) had ALT≥3×ULN. The odds ratio (95% CI) for ALT≥3×ULN for TT genotype was 39.7 (2.2-703.7) compared with other genotypes. As a predictor of ALT≥3×ULN, the TT genotype had a negative predictive value of 0.83 and positive predictive value of 0.67. No TT patients developed liver failure. CONCLUSIONS: The rs2858996/rs707889 polymorphisms in the HFE gene may be associated with reversible ALT elevation in pazo-panib-treated patients with RCC.


Subject(s)
Alanine Transaminase/blood , Antineoplastic Agents/adverse effects , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/enzymology , Histocompatibility Antigens Class I/genetics , Kidney Neoplasms/drug therapy , Kidney Neoplasms/enzymology , Membrane Proteins/genetics , Pyrimidines/adverse effects , Sulfonamides/adverse effects , Aged , Angiogenesis Inhibitors/adverse effects , Carcinoma, Renal Cell/genetics , Female , Genes, MHC Class I , Genes, MHC Class II , Genetic Markers , Hemochromatosis Protein , Humans , Indazoles , Kidney Neoplasms/genetics , Male , Middle Aged , Pharmacogenetics , Polymorphism, Single Nucleotide
9.
J Clin Oncol ; 28(22): 3562-9, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20606083

ABSTRACT

PURPOSE: Pazopanib and lapatinib are tyrosine kinase inhibitors that target vascular endothelial growth factor receptor, platelet-derived growth factor receptor, and c-Kit or epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2/neu), respectively. In cervical cancer, EGFR and HER2/neu overexpression and high microvascular density correlate with survival. PATIENTS AND METHODS: Patients with measurable stage IVB persistent/recurrent cervical carcinoma not amenable to curative therapy and at least one prior regimen in the metastatic setting were randomly assigned in a ratio of 1:1:1 to pazopanib at 800 mg once daily, lapatinib at 1,500 mg once daily, or lapatinib plus pazopanib combination therapy (lapatinib at 1,000 mg plus pazopanib at 400 mg once daily or lapatinib at 1,500 mg plus pazopanib at 800 mg once daily). Therapy continued until progression or withdrawal because of adverse events (AEs). Primary end point was progression-free survival (PFS), and secondary end points were overall survival (OS), response rate (RR), and safety. The futility boundary was crossed at the planned interim analysis for combination therapy compared with lapatinib therapy, and the combination was discontinued. RESULTS: Of 230 patients enrolled, 152 were randomly assigned to the monotherapy arms: pazopanib (n = 74) or lapatinib (n = 78). Most patients (62%) had recurrent cancer. Pazopanib improved PFS (hazard ratio [HR], 0.66; 90% CI, 0.48 to 0.91; P = .013) and OS (HR, 0.67; 90% CI, 0.46 to 0.99; P = .045). Median OS was 50.7 weeks and 39.1 weeks and RRs were 9% and 5% for pazopanib and lapatinib, respectively. The only grade 3 AE > 10% was diarrhea (11% pazopanib and 13% lapatinib). Grade 4 AEs were 9% (lapatinib) and 12% (pazopanib). CONCLUSION: This study confirms the activity of antiangiogenesis agents in advanced and recurrent cervical cancer and demonstrates the benefit of pazopanib based on the prolonged PFS and favorable toxicity profile.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Pyrimidines/administration & dosage , Quinazolines/administration & dosage , Sulfonamides/administration & dosage , Uterine Cervical Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Disease-Free Survival , Drug Delivery Systems , Female , Humans , Indazoles , Lapatinib , Middle Aged , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/adverse effects , Quinazolines/adverse effects , Recurrence , Retreatment , Sulfonamides/adverse effects , Uterine Cervical Neoplasms/mortality
11.
Proc Natl Acad Sci U S A ; 103(51): 19478-83, 2006 Dec 19.
Article in English | MEDLINE | ID: mdl-17164332

ABSTRACT

A critical role for vascular endothelial factor (VEGF) has been demonstrated in multiple myeloma (MM) pathogenesis. Here, we characterized the effect of the small-molecule VEGF receptor inhibitor pazopanib on MM cells in the bone marrow milieu. Pazopanib inhibits VEGF-triggered signaling pathways in both tumor and endothelial cells, thereby blocking in vitro MM cell growth, survival, and migration, and inhibits VEGF-induced up-regulation of adhesion molecules on both endothelial and tumor cells, thereby abrogating endothelial cell-MM cell binding and associated cell proliferation. We show that pazopanib is the first-in-class VEGF receptor inhibitor to inhibit in vivo tumor cell growth associated with increased MM cell apoptosis, decreased angiogenesis, and prolonged survival in a mouse xenograft model of human MM. Low-dose pazopanib demonstrates synergistic cytotoxicity with conventional (melphalan) and novel (bortezomib and immunomodulatory drugs) therapies. Finally, gene expression and signaling network analysis show transcriptional changes of several cancer-related genes, in particular c-Myc. Using siRNA, we confirm the role of c-Myc in VEGF production and secretion, as well as angiogenesis. These preclinical studies provide the rationale for clinical evaluation of pazopanib, alone and in combination with conventional and novel therapies, to increase efficacy, overcome drug resistance, reduce toxicity, and improve patient outcome in MM.


Subject(s)
Endothelial Cells/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Multiple Myeloma/metabolism , Pyrimidines/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction/drug effects , Sulfonamides/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Immunoprecipitation , Indazoles , Mice , Microarray Analysis , Multiple Myeloma/drug therapy , Neovascularization, Pathologic/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Sulfonamides/therapeutic use
12.
Clin Cancer Res ; 12(14 Pt 1): 4265-73, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16857801

ABSTRACT

PURPOSE: Interleukin-18 (IL-18) is an immunostimulatory cytokine with antitumor activity in preclinical animal models. A phase I study of recombinant human IL-18 (rhIL-18) was done to determine the toxicity, pharmacokinetics, and biological activities of rhIL-18 in patients with advanced cancer. EXPERIMENTAL DESIGN: Cohorts of patients were given escalating doses of rhIL-18, each administered as a 2-hour i.v. infusion on 5 consecutive days. Toxicities were graded using standard criteria. Serial blood samples were obtained for pharmacokinetic and pharmacodynamic measurements. RESULTS: Twenty-eight patients (21 with renal cell cancer, 6 with melanoma, and 1 with Hodgkin's lymphoma) were given rhIL-18 in doses ranging from 3 to 1,000 microg/kg. Common side effects included chills, fever, nausea, headache, and hypotension. Common laboratory abnormalities included transient, asymptomatic grade 1 to 2 neutropenia, thrombocytopenia, anemia, hypoalbuminemia, hyponatremia, and elevations in liver transaminases. One patient in the 100 microg/kg cohort experienced transient grade 3 hypotension and grade 2 bradycardia during the first infusion of rhIL-18. No other dose-limiting toxicities were observed. Plasma concentrations of rhIL-18 increased with increasing dose, and 2.5-fold accumulation was observed with repeated dosing. Biological effects of rhIL-18 included transient lymphopenia and increased expression of activation antigens on lymphocytes and monocytes. Increases in serum concentrations of IFN-gamma, granulocyte macrophage colony-stimulating factor, IL-18 binding protein, and soluble Fas ligand were observed. Two patients experienced unconfirmed partial responses after rhIL-18 treatment. CONCLUSIONS: rhIL-18 can be safely given in biologically active doses to patients with advanced cancer. A maximum tolerated dose of rhIL-18 was not determined. Further clinical studies of rhIL-18 are warranted.


Subject(s)
Interleukin-18/administration & dosage , Neoplasms/drug therapy , Neoplasms/metabolism , Recombinant Proteins/administration & dosage , Adult , Aged , Area Under Curve , Carcinoma, Renal Cell/drug therapy , Cohort Studies , Cytokines/metabolism , Dose-Response Relationship, Drug , Female , Hodgkin Disease/drug therapy , Humans , Infusions, Intravenous , Male , Melanoma/drug therapy , Middle Aged
13.
Blood ; 103(9): 3474-9, 2004 May 01.
Article in English | MEDLINE | ID: mdl-14644994

ABSTRACT

Previous studies have shown that the multiple myeloma (MM) cell line and MM patient cells express high-affinity vascular endothelial growth factor (VEGF) receptor-1 or Fms-like tyrosine kinase-1 (Flt-1) but not VEGF receptor-2 or Flk-1/kinase insert domain-containing receptor (Flk-1/KDR) and that VEGF triggers MM cell proliferation through a mitogen-activated protein kinase (MAPK)-dependent pathway and migration through a protein kinase C (PKC)-dependent pathway. The present study evaluates the efficacy of the small molecule tyrosine-kinase inhibitor GW654652, which inhibits all 3 VEGF receptors with similar potency. We show that GW654652 acts directly on MM cells and in the bone marrow microenvironment. Specifically, GW654652 (1-10 microg/mL) inhibits, in a dose-dependent fashion, VEGF-triggered migrational activity and cell proliferation of MM cell lines that are sensitive and resistant to conventional therapy. As expected from our previous studies of VEGF-induced signaling and sequelae in MM cells, GW654652 blocked VEGF-induced Flt-1 phosphorylation and downstream activation of AKT-1 and MAPK-signaling cascades. Importantly, GW654652 also inhibits interleukin-6 and VEGF secretion and proliferation of MM cells induced by tumor cell binding to bone marrow (BM) stromal cells. The activity of a pan-VEGF receptor inhibitor against MM cells in the BM milieu, coupled with its lack of major toxicity in preclinical mouse models, provides the framework for clinical trials of this drug class to improve patient outcome in MM.


Subject(s)
Multiple Myeloma/pathology , Proto-Oncogene Proteins , Pyrimidines/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Bone Marrow/pathology , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Interleukin-6/metabolism , Multiple Myeloma/drug therapy , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...