Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Clin Transl Sci ; 17(3): e13730, 2024 03.
Article in English | MEDLINE | ID: mdl-38411318

ABSTRACT

Like other monoclonal antibodies, immune checkpoint inhibitors may be immunogenic in some patients, potentially affecting pharmacokinetics (PKs) and clinical outcomes. In post hoc analyses, we characterized antidrug antibody (ADA) development with avelumab monotherapy in patients with metastatic Merkel cell carcinoma (mMCC) from the JAVELIN Merkel 200 trial (first-line [1L; N = 116] and second-line or later [≥2L; N = 88] cohorts) or with advanced urothelial carcinoma (aUC) from the JAVELIN Bladder 100 (1L maintenance [N = 350]) and JAVELIN Solid Tumor (≥2L [N = 249]) trials. Treatment-emergent ADAs developed in a numerically higher proportion of patients with aUC (1L maintenance, 19.1%; ≥2L, 18.1%) versus mMCC (1L, 8.2%; ≥2L, 8.9%); incidences within tumor types were similar by line of therapy. In PK analyses, numerically lower avelumab trough concentration and higher baseline clearance were observed in treatment-emergent ADA+ versus ADA- subgroups; however, differences were not clinically relevant. Numerical differences in overall survival, progression-free survival, or objective response rate by ADA status were observed; however, no clinically meaningful trends were identified. Proportions of patients with treatment-emergent adverse events (TEAEs; any grade or grade 3/4), serious TEAEs, TEAEs leading to treatment discontinuation, or infusion-related reactions were similar, with overlapping 80% confidence intervals between ADA subgroups. Efficacy and safety observations were similar in subgroups defined by early development of ADA+ status during treatment. In conclusion, no meaningful differences in PKs, efficacy, and safety were observed between subgroups of avelumab-treated patients with different ADA status. Overall, these data suggest that ADAs are not relevant for treatment decisions with avelumab.


Subject(s)
Carcinoma, Merkel Cell , Carcinoma, Transitional Cell , Skin Neoplasms , Urinary Bladder Neoplasms , Humans , Antibodies, Monoclonal, Humanized/adverse effects , Carcinoma, Merkel Cell/drug therapy , Carcinoma, Merkel Cell/pathology , Carcinoma, Transitional Cell/drug therapy , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Clinical Trials as Topic
2.
CPT Pharmacometrics Syst Pharmacol ; 12(12): 2001-2012, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37794707

ABSTRACT

Exposure-response (E-R) analyses are an integral component of understanding the benefit/risk profile of novel oncology therapeutics. These analyses are typically conducted using data from the treatment arm to characterize the relationship between drug exposure (low vs. high) and efficacy or safety outcomes. For example, outcomes of patients with lower exposure in the treatment arm (e.g., Q1) might be compared to outcomes of those with higher drug exposure (Q2, Q3, and Q4). Outcomes from the lowest exposure quartile may be also compared to the control arm to evaluate whether the Q1 subgroup derived clinical benefit. However, the sample size and the distribution of patient baseline characteristics and disease risk factors are not balanced in such a comparison (Q1 vs. control), which may bias the analysis and causal interpretation of clinical benefit in the Q1 subgroup. Herein, we report the use of case-control matching to account for this bias and better understand the E-R relationship for avelumab in urothelial carcinoma, a PD-L1 inhibitor approved for the treatment of several cancers. Data from JAVELIN-100 was utilized which is a phase III study of avelumab in first-line maintenance treatment in patients with urothelial carcinoma; this clinical study demonstrated superiority of avelumab versus best-supportive care leading to approval in the United States, Europe, and other countries. A post hoc case-control matching method was implemented to compare the efficacy outcome between Q1 avelumab subgroup and matched patients extracted from the control arm with similar baseline characteristics, which showed a clinically relevant difference in overall survival in favor of the Q1 avelumab subgroup. This analysis demonstrates the importance of accounting for imbalance in important baseline covariates when comparing efficacy outcomes between subgroups within the treatment arm versus the control arm.


Subject(s)
Carcinoma, Transitional Cell , Urinary Bladder Neoplasms , Humans , Antibodies, Monoclonal/therapeutic use , Carcinoma, Transitional Cell/drug therapy , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/chemically induced , Case-Control Studies
3.
Clin Pharmacol Ther ; 107(3): 588-596, 2020 03.
Article in English | MEDLINE | ID: mdl-31553054

ABSTRACT

Avelumab, an anti-programmed death-ligand 1 monoclonal antibody approved for the treatment of metastatic Merkel cell carcinoma and platinum-treated urothelial carcinoma, was initially approved with a 10 mg/kg weight-based dose. We report pharmacokinetic (PK)/pharmacodynamic analyses for avelumab comparing weight-based dosing and a flat 800 mg dose, developed using data from 1,827 patients enrolled in 3 clinical trials (NCT01772004, NCT01943461, and NCT02155647). PK metrics were simulated for weight-based and flat-dosing regimens and summarized by quartiles of weight. Derived exposure metrics were used in simulations of exposure-safety (various tumors) and exposure-efficacy (objective responses; Merkel cell or urothelial carcinoma). Flat dosing was predicted to provide similar exposure to weight-based dosing, with slightly lower variability. Exposure-safety and exposure-efficacy simulations suggested similar benefit:risk profiles for the two dosing regimens. These pharmacometric analyses provided the basis for the US Food and Drug Administration approval of a flat dose of avelumab 800 mg every 2 weeks in approved indications.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Carcinoma, Merkel Cell/drug therapy , Skin Neoplasms/drug therapy , Urologic Neoplasms/drug therapy , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacology , Body Weight , Clinical Trials as Topic , Computer Simulation , Dose-Response Relationship, Drug , Humans
4.
CPT Pharmacometrics Syst Pharmacol ; 8(6): 415-427, 2019 06.
Article in English | MEDLINE | ID: mdl-30980481

ABSTRACT

Avelumab, a human anti-programmed death ligand 1 immunoglobulin G1 antibody, has shown efficacy and manageable safety in multiple tumors. A two-compartment population pharmacokinetic model for avelumab incorporating intrinsic and extrinsic covariates and time-varying clearance (CL) was identified based on data from 1,827 patients across three clinical studies. Of 14 tumor types, a decrease in CL over time was more notable in metastatic Merkel cell carcinoma and squamous cell carcinoma of the head and neck, which had maximum decreases of 32.1% and 24.7%, respectively. The magnitude of reduction in CL was higher in responders than in nonresponders. Significant covariate effects of baseline weight, baseline albumin, and sex were identified on both CL and central distribution volume. Significant covariate effects of black/African American race, C-reactive protein, and immunogenicity were found on CL. None of the covariate or time-dependent effects were clinically important or warranted dose adjustment.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Carcinoma, Merkel Cell/drug therapy , Carcinoma, Transitional Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Skin Neoplasms/drug therapy , Squamous Cell Carcinoma of Head and Neck/drug therapy , Adult , Aged , Aged, 80 and over , Carcinoma, Merkel Cell/metabolism , Carcinoma, Transitional Cell/metabolism , Clinical Trials as Topic , Female , Head and Neck Neoplasms/metabolism , Humans , Male , Metabolic Clearance Rate , Middle Aged , Serum Albumin/metabolism , Sex Factors , Skin Neoplasms/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism , Time Factors , Treatment Outcome , Young Adult
5.
J Clin Pharmacol ; 56(2): 223-30, 2016 02.
Article in English | MEDLINE | ID: mdl-26179237

ABSTRACT

This phase 1, open-label crossover study evaluated the relative bioavailability of dacomitinib in healthy volunteers under fed and fasted conditions and following coadministration with rabeprazole, a potent acid-reducing proton pump inhibitor (PPI). Twenty-four male subjects received a single dacomitinib 45-mg dose under 3 different conditions separated by washout periods of ≥ 16 days: coadministered with rabeprazole 40 mg under fasting conditions; alone under fasting conditions; and alone after a high-fat, high-calorie meal. Increased peak exposure of 23.7% (90% confidence interval [CI], 5.3%-45.2%) was detected with dacomitinib taken after food versus fasting. The adjusted geometric mean ratio (fed/fasted) for area under the plasma concentration-time curve from time zero to infinity (AUCinf ) was 114.2% (90%CI, 104.7%-124.5%) and not considered clinically meaningful. In the fasted state, a decrease in dacomitinib AUCinf was observed following rabeprazole versus dacomitinib alone (PPI+fasted/fasted alone): 71.1% (90%CI, 61.7%-81.8%). Dacomitinib was generally well tolerated. Dacomitinib may be taken with or without food. Use of long-acting acid-reducing agents, such as PPIs with dacomitinib should be avoided if possible. Shorter-acting agents such as antacids and H2-receptor antagonists may have lesser impact on dacomitinib exposure and may be preferable to PPIs if acid reduction is clinically required.


Subject(s)
Drug Antagonism , Food-Drug Interactions , Quinazolinones/pharmacokinetics , Rabeprazole/pharmacokinetics , Adult , Area Under Curve , Biological Availability , Cross-Over Studies , Dietary Fats , Energy Intake , Female , Half-Life , Humans , Male , Middle Aged , Proton Pump Inhibitors/administration & dosage , Proton Pump Inhibitors/pharmacokinetics , Quinazolinones/administration & dosage , Rabeprazole/administration & dosage , Young Adult
6.
Invest New Drugs ; 33(4): 931-41, 2015 08.
Article in English | MEDLINE | ID: mdl-26048096

ABSTRACT

Dacomitinib (PF-00299804) is a small-molecule inhibitor of the tyrosine kinases human epidermal growth factor receptor-1 (HER1; epidermal growth factor receptor, EGFR), HER2, and HER4 currently being developed for the treatment of lung cancer with sensitizing mutations in EGFR or refractory to EGFR-directed treatment. Dacomitinib is largely metabolized by the liver through oxidative and conjugative metabolism; therefore, determination of the impact of varying degrees of hepatic impairment on the pharmacokinetics (PK) of dacomitinib was warranted to ensure patient safety. In this phase I, open-label, parallel-group study, a single dose of dacomitinib was administered to healthy volunteers and to subjects with mild or moderate liver dysfunction, as determined by Child-Pugh classification. The primary goal of this study was to evaluate the effects of mild and moderate hepatic impairment on the single-dose PK profile of dacomitinib, as well as to assess the safety and tolerability in these subjects. Plasma protein binding and impact of hepatic function on the PK of the active metabolite PF-05199265 was also investigated. Twenty-five male subjects received dacomitinib 30 mg, with 8 subjects in the healthy- and mild-impairment cohorts and 9 subjects in the moderate-impairment cohort. Compared with healthy volunteers, there was no significant change in dacomitinib exposure in subjects with mild or moderate liver dysfunction and no observed alteration in plasma protein binding. No serious treatment-related adverse events were reported in any group, and dacomitinib was well tolerated. A dose adjustment does not appear necessary when administering dacomitinib to patients with mild or moderate hepatic impairment.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Liver Diseases/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Quinazolinones/pharmacokinetics , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Blood Proteins/metabolism , Cytochrome P-450 CYP2D6/genetics , ErbB Receptors/antagonists & inhibitors , Genotype , Humans , Male , Middle Aged , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/blood , Quinazolinones/adverse effects , Quinazolinones/blood
7.
J Clin Pharmacol ; 54(5): 555-62, 2014 May.
Article in English | MEDLINE | ID: mdl-24293056

ABSTRACT

Dacomitinib is currently in development for the treatment of non-small cell lung cancer. Formation of the major circulating metabolite (PF-05199265) is mediated by cytochrome P450 (CYP) 2D6 and CYP2C9. This phase I, single fixed-sequence, two-period study evaluated the effect of paroxetine, a CYP2D6 inactivator, on dacomitinib pharmacokinetics in healthy volunteers who were extensive CYP2D6 metabolizers. Subjects received a single 45-mg dacomitinib dose alone and in combination with paroxetine (30 mg/day for 10 consecutive days, with dacomitinib administered on day 4) at steady-state levels. Blood samples were collected through 240 hours post-dacomitinib dosing. Dacomitinib exposure (area under the concentration-time curve from 0 to infinity; AUCinf) increased 37%; however a reduction in PF-05199265 AUCinf of approximately 90% was observed during the paroxetine treatment period. The maximum concentration of dacomitinib changed minimally. Adverse events reported with single-dose dacomitinib administered alone or in the presence of steady-state levels of paroxetine were mostly mild, and no serious adverse events were reported. While paroxetine significantly inhibited CYP2D6-mediated metabolism of a single dose of dacomitinib, the modest effect on dacomitinib exposure is unlikely to be clinically relevant when dacomitinib is given daily. Dose adjustment of dacomitinib may therefore not be required upon coadministration with a CYP2D6 inhibitor.


Subject(s)
Cytochrome P-450 CYP2D6 Inhibitors/pharmacology , Paroxetine/pharmacology , Quinazolinones/pharmacokinetics , Adult , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP2D6 Inhibitors/adverse effects , Cytochrome P-450 CYP2D6 Inhibitors/blood , Cytochrome P-450 CYP2D6 Inhibitors/pharmacokinetics , Drug Interactions , Healthy Volunteers , Humans , Male , Middle Aged , Paroxetine/adverse effects , Paroxetine/blood , Paroxetine/pharmacokinetics , Quinazolinones/adverse effects , Quinazolinones/blood , Young Adult
8.
Cancer Chemother Pharmacol ; 72(2): 379-85, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23760812

ABSTRACT

PURPOSE: This study aimed to characterize the primary routes of elimination of the pan-HER tyrosine kinase inhibitor, dacomitinib (PF-00299804), to evaluate the pharmacokinetics of total radioactivity and of dacomitinib and to identify the metabolites of dacomitinib in plasma, urine, and feces in the healthy volunteers. METHODS: Six male healthy volunteers (mean age 31.5 years) received a single 45-mg oral dose containing ~100 µCi [(14)C] dacomitinib. Whole blood, urine, and fecal samples were collected throughout the study and analyzed for total radioactivity by liquid scintillation counting. Safety evaluations included vital signs, 12-lead ECGs, safety laboratory tests, and monitoring of adverse events. RESULTS: 78.8 % of the radiolabeled material was excreted in feces, and 3.2 % was recovered in urine. Peak concentrations of dacomitinib in plasma occurred 12 h (median) after oral dosing. Mean terminal plasma half-life was 55 and 182 h for dacomitinib and total plasma radioactivity, respectively. Geometric mean C max was approximately 2-fold higher, and total exposure (AUCinf) was almost 6-fold higher for total radioactivity than for dacomitinib in plasma. O-desmethyl dacomitinib (PF-05199265) was the major circulating metabolite. T max of this metabolite occurred 6 h after oral dosing with dacomitinib. Plasma exposure for the metabolite was one-third that of the parent compound. There were no serious/severe adverse events or deaths during the study. Dacomitinib was well tolerated. CONCLUSIONS: In humans, [(14)C] dacomitinib underwent oxidative and conjugative metabolism. Most of the administered dose was eliminated via the fecal route, and the major circulating metabolite was PF-05199265.


Subject(s)
Protein Kinase Inhibitors/pharmacokinetics , Quinazolinones/pharmacokinetics , Adult , Area Under Curve , Biotransformation , Carbon Radioisotopes , Chromatography, High Pressure Liquid , Electrocardiography/drug effects , Feces/chemistry , Half-Life , Humans , Isotope Labeling , Male , Mass Spectrometry , Middle Aged , Protein Kinase Inhibitors/adverse effects , Quinazolinones/adverse effects , Young Adult
9.
Drug Metab Dispos ; 40(3): 539-55, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22180047

ABSTRACT

Sunitinib is an oral multitargeted tyrosine kinase inhibitor approved for the treatment of advanced renal cell carcinoma, imatinib-refractory gastrointestinal stromal tumor, and advanced pancreatic neuroendocrine tumors. The current studies were conducted to characterize the pharmacokinetics, distribution, and metabolism of sunitinib after intravenous and/or oral administrations of [(14)C]sunitinib in rats (5 mg/kg i.v., 15 mg/kg p.o.), monkeys (6 mg/kg p.o.), and humans (50 mg p.o.). After oral administration, plasma concentration of sunitinib and total radioactivity peaked from 3 to 8 h. Plasma terminal elimination half-lives of sunitinib were 8 h in rats, 17 h in monkeys, and 51 h in humans. The majority of radioactivity was excreted to the feces with a smaller fraction of radioactivity excreted to urine in all three species. The bioavailability in female rats was close to 100%, suggesting complete absorption of sunitinib. Whole-body autoradioluminography suggested radioactivity was distributed throughout rat tissues, with the majority of radioactivity cleared within 72 h. Radioactivity was eliminated more slowly from pigmented tissues. Sunitinib was extensively metabolized in all species. Many metabolites were detected both in urine and fecal extracts. The main metabolic pathways were N-de-ethylation and hydroxylation of indolylidene/dimethylpyrrole. N-Oxidation/hydroxylation/desaturation/deamination of N,N'-diethylamine and oxidative defluorination were the minor metabolic pathways. Des-ethyl metabolite M1 was the major circulating metabolite in all three species.


Subject(s)
Indoles/metabolism , Indoles/pharmacokinetics , Pyrroles/metabolism , Pyrroles/pharmacokinetics , Absorption/physiology , Administration, Oral , Adult , Animals , Biological Availability , Carbon Isotopes/administration & dosage , Carbon Isotopes/metabolism , Carbon Isotopes/urine , Carbon Radioisotopes , Feces/chemistry , Female , Half-Life , Humans , Indoles/administration & dosage , Indoles/urine , Injections, Intravenous/methods , Macaca fascicularis , Male , Middle Aged , Pyrroles/administration & dosage , Pyrroles/urine , Rats , Rats, Sprague-Dawley , Sunitinib , Tissue Distribution , Young Adult
10.
Cancer Chemother Pharmacol ; 69(4): 991-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22147075

ABSTRACT

PURPOSE: This study evaluated the effect of a single 45-mg dose of dacomitinib (PF-00299804), an irreversible small-molecule inhibitor of human epidermal growth factor receptors-1, -2, and -4, on CYP2D6 activity in healthy volunteers (HV) using dextromethorphan (DM), a selective CYP2D6 probe. METHODS: Fourteen male HVs were enrolled in this open-label, randomized, cross-over, single-dose study of DM alone or with dacomitinib. Each HV received both treatments separated by a 14-day washout period. The pharmacokinetics of DM, dextrorphan (DX; the major DM metabolite), dacomitinib and PF-05199265 (an active metabolite of dacomitinib) were calculated. RESULTS: When combined with dacomitinib, the ratio of adjusted geometric means (90% CI) of DM area under the concentration-time curve (AUC)(last) was 955% (90% CI: 560%, 1,630%) and maximum plasma concentration (C (max)) was 973% (90% CI: 590%, 1,606%), compared with DM alone. For dacomitinib plus DM, exposures were consistent with those in patients receiving single-dose dacomitinib. Terminal elimination half-life (t (1/2)) was 51.4 h. Mild and moderate treatment-related adverse events were reported. No HV withdrew from the study. CONCLUSIONS: Single-dose administration of dacomitinib plus DM was safe and well tolerated in HVs and resulted in a significant increase in systemic exposures of DM in extensive metabolizers. No effect was observed on the pharmacokinetics of dacomitinib. Drug-drug interaction may occur when dacomitinib is concomitantly administered with therapeutic agents metabolized by cytochrome P450 (CYP) 2D6. Administration of drugs which are highly dependent on CYP2D6 metabolism may require dose adjustment, or substitution with an alternative medication.


Subject(s)
Cytochrome P-450 CYP2D6 Inhibitors , Dextromethorphan/pharmacology , Quinazolinones/pharmacology , Adult , Cross-Over Studies , Cytochrome P-450 CYP2D6/metabolism , Dextromethorphan/adverse effects , Dextromethorphan/pharmacokinetics , Drug Interactions , Humans , Male , Quinazolinones/adverse effects , Quinazolinones/pharmacokinetics
11.
Cell Immunol ; 271(2): 299-307, 2011.
Article in English | MEDLINE | ID: mdl-21872226

ABSTRACT

Thymosin-ß4 (Tß4) is a G-actin sequestering peptide involved in regeneration and remodeling of injured tissues. In this work, we have designed and synthesized three peptide sequences containing the N-terminus (TYB4-n), the central part (TYB4-i) or the C-terminus (TYB4-c) of Tß4. All fragments are overlapping on the main central binding actin site. After a structural characterization, we have evaluated in vitro and in vivo their pro-angiogenic effects. The results of this study have shown that: (i) each fragment reproduces the native conformation; (ii) Tß4-derived peptides exert both in vitro and in vivo pro-angiogenic effects; (iii) their in vitro effect seem to be related to the activation of several signaling pathways and is positively modulated by the N-terminus of Tß4.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Neovascularization, Physiologic/drug effects , Thymosin/pharmacology , Amino Acid Sequence , Angiogenesis Inducing Agents/chemistry , Animals , Cell Proliferation/drug effects , Chick Embryo , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Models, Molecular , Molecular Sequence Data , Neovascularization, Physiologic/physiology , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Protein Conformation , Signal Transduction/drug effects , Thymosin/chemistry , Thymosin/genetics , Thymosin/physiology , Wound Healing/drug effects , Wound Healing/physiology
12.
Anticancer Drugs ; 22(5): 377-83, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21394020

ABSTRACT

Targeted agents such as sunitinib, an oral, multitargeted receptor tyrosine kinase inhibitor, have greatly improved the prognosis for patients with metastatic renal cell carcinoma (mRCC). In this review we analyse data from sunitinib preclinical and clinical studies in detail and consider the key implications for the effective use of sunitinib in clinical practice. Sunitinib has shown efficacy and acceptable tolerability in patients with mRCC in phase II and III clinical studies. In a pivotal phase III study in treatment-naïve patients with mRCC, median progression-free survival for sunitinib-treated patients was double of that with interferon-α (P < 0.001). Median overall survival was 26.4 versus 21.8 months, respectively (P = 0.0510). In preclinical and phase I/II studies, sunitinib inhibits tyrosine kinase inhibitors in a dose-dependent manner, suggesting a correlation between increasing exposure and greater response. A pharmacokinetics/pharmacodynamics meta-analysis investigating the relationship between clinical end points and sunitinib exposure showed that increased sunitinib exposure was associated with a greater probability of objective response, longer time to tumour progression and overall survival, as well as some increased risk of specific adverse events. It is important to consider the relationship between exposure and response to maximize clinical benefit from sunitinib treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Renal Cell/drug therapy , Indoles/administration & dosage , Kidney Neoplasms/drug therapy , Pyrroles/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Carcinoma, Renal Cell/metabolism , Clinical Trials as Topic , Humans , Indoles/adverse effects , Indoles/pharmacokinetics , Kidney Neoplasms/metabolism , Meta-Analysis as Topic , Neoplasm Metastasis , Pyrroles/adverse effects , Pyrroles/pharmacokinetics , Sunitinib
13.
J Pept Sci ; 16(7): 349-57, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20552562

ABSTRACT

It is well known that tumor growth is strictly dependent on neo-vessel formation inside the tumor mass and that cell adhesion is required to allow EC proliferation and migration inside the tumor. In this work, we have evaluated the in vitro and in vivo effects on angiogenesis of some peptides, originally designed to promote cell adhesion on biomaterials, containing RGD motif mediating cell adhesion via integrin receptors [RGD, GRGDSPK, and (GRGDSP)(4)K] or the heparin-binding sequence of human vitronectin that interacts with HSPGs [HVP(351-359)]. Cell adhesion, proliferation, migration, and capillary-like tube formation in Matrigel were determined on HUVECs, whereas the effects on in vivo angiogenesis were evaluated using the CAM assay. (GRGDSP)(4)K linear sequence inhibited cell adhesion, decreased cell proliferation, migration and morphogenesis in Matrigel, and induced anti-angiogenic responses on CAM at higher degree than that determined after incubation with RGD or GRGDSPK. Moreover, it counteracted both in vitro and in vivo the pro-angiogenic effects induced by the Fibroblast growth factor (FGF-2). On the other hand, HVP was not able to affect cell adhesion and appeared less effective than (GRGDSP)(4)K. Our data indicate that the activity of RGD-containing peptides is related to their adhesive properties, and their effects are modulated by the number of cell adhesion motifs and the aminoacidic residues next to these sequences. The anti-angiogenic properties of (GRGDSP)(4)K seem to depend on its interaction with integrins, whereas the effects of HVP may be partially due to an impairment of HSPGs/FGF-2.


Subject(s)
Cell Adhesion/drug effects , Endothelium, Vascular/cytology , Neovascularization, Physiologic/drug effects , Oligopeptides/pharmacology , Amino Acid Sequence , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Humans , Integrins , Oligopeptides/chemistry
14.
Cancer Chemother Pharmacol ; 66(4): 699-707, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20049443

ABSTRACT

This study evaluated the effect of hepatic impairment on the pharmacokinetics of sunitinib and its active metabolite, SU12662. This open-label study enrolled subjects with normal hepatic function (n = 8), mild (Child-Pugh [CP]-A; n = 8), or moderate (CP-B; n = 8) hepatic impairment. Subjects received sunitinib 50 mg as a single oral dose. Mild or moderate hepatic impairment did not significantly alter sunitinib, SU12662, or total drug (TD) systemic exposure. In subjects with normal hepatic function, mild, or moderate hepatic impairment, respectively, TD AUC(0-infinity) was 1,938, 2,002, and 1,999 ng h/ml, TD AUC(0-last) was 1,913, 1,956, and 1,958 ng h/ml, and TD C (max) was 26.0, 27.3, and 26.7 ng/ml. There were no other notable pharmacokinetic differences and sunitinib was well tolerated. The pharmacokinetic findings of this study do not indicate a need to adjust the currently approved starting dose of sunitinib (50 mg daily on Schedule 4/2) for cancer patients with mild to moderate liver impairment.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Indoles/pharmacokinetics , Liver Diseases/metabolism , Pyrroles/pharmacokinetics , Aged , Alanine Transaminase/blood , Antineoplastic Agents/adverse effects , Area Under Curve , Endpoint Determination , Female , Humans , Indoles/adverse effects , Liver Function Tests , Male , Middle Aged , Pyrroles/adverse effects , Sample Size , Sunitinib
15.
Cancer Chemother Pharmacol ; 66(2): 357-71, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19967539

ABSTRACT

PURPOSE: In this pharmacokinetic/pharmacodynamic meta-analysis, we investigated relationships between clinical endpoints and sunitinib exposure in patients with advanced solid tumors, including patients with gastrointestinal stromal tumor (GIST) and metastatic renal cell carcinoma (mRCC). METHODS: Pharmacodynamic data were available for 639 patients of whom 443 had pharmacokinetic data. Sunitinib doses ranged from 25 to 150 mg QD or QOD. Models to express endpoint values and/or changes from baseline by the highest-correlating exposure measures were developed in S-PLUS or NONMEM using fixed- and mixed-effects modeling. RESULTS: Tentative relationships were identified between (1) steady-state AUC of total drug (sunitinib + its active metabolite SU12662) and time to tumor progression (TTP), overall survival (OS), with AUC significantly associated with longer TTP and OS in patients with GIST and mRCC, and incidence, but not severity, of fatigue; (2) steady-state AUC of sunitinib and response probability, with AUC significantly associated with objective response in patients with mRCC and stable disease in patients with both mRCC and GIST (with no such correlations in patients with solid tumors); (3) dose and tumor size reductions; (4) total drug concentration and diastolic blood pressure (DBP), with a typical patient on sunitinib 50 mg QD (the recommended dose) predicted to experience a maximum DBP increase of 8 mmHg; and (5) cumulative AUC of total drug and absolute neutrophil count (ANC), with ANC reductions occurring predominantly after one treatment cycle. CONCLUSIONS: The results of this meta-analysis indicate that increased exposure to sunitinib is associated with improved clinical outcomes (longer TTP, longer OS, greater chance of antitumor response), as well as some increased risk of adverse effects. A sunitinib 50-mg starting dose seems reasonable, providing clinical benefit with acceptably low risk of adverse events.


Subject(s)
Antineoplastic Agents/therapeutic use , Indoles/therapeutic use , Neoplasms/drug therapy , Pyrroles/therapeutic use , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Blood Pressure/drug effects , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Endpoint Determination , Fatigue/chemically induced , Fatigue/epidemiology , Female , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/pathology , Humans , Indoles/adverse effects , Indoles/pharmacokinetics , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Leukocyte Count , Linear Models , Male , Middle Aged , Neutrophils/drug effects , Nonlinear Dynamics , Population , Pyrroles/adverse effects , Pyrroles/pharmacokinetics , Sunitinib
16.
J Clin Pharmacol ; 50(4): 472-81, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19779038

ABSTRACT

This phase I, open-label, single-dose study evaluates the effects of severe renal impairment and end-stage renal disease (ESRD) requiring hemodialysis on the pharmacokinetics, safety, and tolerability of sunitinib and its primary active metabolite, SU12662. Subjects with normal renal function (creatinine clearance > 80 mL/min), severe renal impairment (creatinine clearance < 30 mL/min), and ESRD requiring hemodialysis receive a single dose of sunitinib 50 mg. Serial blood samples are collected for quantification of plasma concentrations using a validated liquid chromatography with tandem mass spectrometry assay. Safety is monitored. Twenty-four subjects complete the study. Pharmacokinetics in subjects with severe renal impairment appear similar to those with normal renal function. Plasma exposure to sunitinib and SU12662 appears lower in subjects with ESRD compared with subjects with normal renal function or severe renal impairment. Single-dose sunitinib 50 mg is well tolerated regardless of renal function. The currently approved starting dose of sunitinib 50 mg on Schedule 4/2 is expected to be appropriate for patients with renal impairment; any subsequent dose modifications should be based on patients' ability to tolerate treatment.


Subject(s)
Indoles/adverse effects , Indoles/pharmacokinetics , Kidney Failure, Chronic/drug therapy , Kidney Failure, Chronic/physiopathology , Kidney Function Tests , Pyrroles/adverse effects , Pyrroles/pharmacokinetics , Adult , Aged , Female , Follow-Up Studies , Humans , Kidney Failure, Chronic/metabolism , Male , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/physiology , Middle Aged , Sunitinib
17.
Clin Cancer Res ; 15(22): 7045-52, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19903787

ABSTRACT

PURPOSE: To evaluate the effects of sunitinib, a multitargeted tyrosine kinase inhibitor, on the QT interval in patients with cancer. EXPERIMENTAL DESIGN: Patients received sunitinib loading doses (150-200 mg) on days 3 and 9 and maintenance doses (50 mg/d) on days 4 to 8. Moxifloxacin (day 1), placebo (day 2), and granisetron [with placebo (day 2) or sunitinib (days 3 and 9)] were also administered. Treatment effects were evaluated by time-matched, serial electrocardiograms, and manually overread. RESULTS: Twenty-four of 48 patients were QT/PK evaluable. Moxifloxacin produced a time-matched, maximum mean placebo-adjusted corrected QT interval (QT(c)F) of 5.6 ms [90% confidence interval (CI), 1.9-9.3]. Sunitinib QT(c)F changes correlated with exposure, but not T(max). Maximum mean time-matched, placebo-adjusted QT(c)F was 9.6 ms (90% CI, 4.1-15.1) at steady state/therapeutic concentrations (day 3) and 15.4 ms (90% CI, 8.4-22.4) at supratherapeutic concentrations (day 9). No patient had a QT(c)F >500 ms. Concomitant granisetron produced no significant QT(c)F prolongation. Sunitinib-related adverse events were as previously described. CONCLUSIONS: Sunitinib has a dose-dependent effect on QT interval. The increased risk of ventricular arrhythmias must be weighed against the therapeutic benefit sunitinib provides to patients with advanced cancer.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Arrhythmias, Cardiac/complications , Electrocardiography/methods , Indoles/pharmacokinetics , Neoplasms/complications , Neoplasms/drug therapy , Pyrroles/pharmacokinetics , Antineoplastic Agents/pharmacology , Arrhythmias, Cardiac/chemically induced , Aza Compounds/therapeutic use , Dose-Response Relationship, Drug , Fluoroquinolones , Granisetron/therapeutic use , Heart Ventricles/pathology , Humans , Indoles/pharmacology , Moxifloxacin , Placebos , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrroles/pharmacology , Quinolines/therapeutic use , Risk , Sunitinib , Time Factors
18.
Clin Cancer Res ; 15(18): 5902-9, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19737946

ABSTRACT

PURPOSE: To evaluate sunitinib activity and potential cellular and molecular correlates in gastrointestinal stromal tumor (GIST) patients after imatinib failure, in addition to assessing the safety and pharmacokinetics (PK) of different dose schedules. EXPERIMENTAL DESIGN: In this open-label, dose-ranging, phase I/II study, 97 patients with metastatic imatinib-resistant/intolerant GIST received sunitinib at doses of 25, 50, or 75 mg/d on one of three schedules. Serial tumor imaging was done using computed tomography and [18F]fluoro-2-deoxy-d-glucose positron emission tomography scanning. PK and cell proliferation and KIT phosphorylation status in tumor biopsies were also analyzed. RESULTS: Clinical benefit was observed in 52 patients (54%: 7 objective partial responses, 45 stable disease > or =6 months). Decreased tumor glycolytic activity was shown in most patients within 7 days of starting sunitinib using [18F]fluoro-2-deoxy-d-glucose positron emission tomography. Sunitinib treatment was associated with reduced tumor cell proliferation by >25% in 52% of cases analyzed and reduced levels of phospho-KIT in tumor biopsies (indicating target modulation). The recommended dose schedule was 50 mg/d for 4 weeks followed by 2 weeks off treatment. On the 50-mg dose across all schedules, 79% of PK-evaluable patients achieved total drug trough concentrations above the target concentration (50 ng/mL) within 14 days of dosing. In addition, adverse events were generally mild to moderate in severity. CONCLUSION: Cellular and molecular analyses showed that sunitinib clinical activity is associated with inhibition of KIT in GIST following imatinib failure, illustrating the rational approach used to develop a therapy aimed at the underlying oncogenic signaling pathway aberrancy.


Subject(s)
Gastrointestinal Stromal Tumors/diagnosis , Gastrointestinal Stromal Tumors/drug therapy , Indoles/administration & dosage , Indoles/therapeutic use , Piperazines/therapeutic use , Positron-Emission Tomography , Pyrimidines/therapeutic use , Pyrroles/administration & dosage , Pyrroles/therapeutic use , Adult , Aged , Benzamides , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Fluorodeoxyglucose F18 , Gastrointestinal Stromal Tumors/secondary , Humans , Imatinib Mesylate , Indoles/pharmacology , Male , Middle Aged , Piperazines/administration & dosage , Piperazines/pharmacology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Pyrroles/pharmacology , Sunitinib , Treatment Failure
19.
Clin Cancer Res ; 15(7): 2497-506, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19258444

ABSTRACT

PURPOSE: Sunitinib malate is an oral multitargeted tyrosine kinase inhibitor approved for advanced renal cell carcinoma and imatinib-resistant or imatinib-intolerant gastrointestinal stromal tumor. Following administration, sunitinib is metabolized by cytochrome P450 3A4 to an active metabolite (SU12662). The objective of this analysis was to assess sunitinib and SU12662 pharmacokinetics and to identify covariates that might explain variability in exposure following oral administration. EXPERIMENTAL DESIGN: Data from 590 subjects (73 volunteers and 517 patients) in 14 studies were analyzed. Plasma concentration-time data were analyzed using nonlinear mixed-effects modeling to estimate population pharmacokinetic parameters, as well as relationships between these parameters and gender, race, age, weight, creatinine clearance, Eastern Cooperative Oncology Group score, and tumor type. Simulations were done to determine the predicted effect of these covariates on exposure. RESULTS: Separate models were developed for sunitinib and SU12662 (each a two-compartment model with first-order absorption and elimination). Sunitinib parameters were estimated as CL/F, 51.8 L/h and Vd/F(central), 2,030 liters. SU12662 parameters were estimated as CL/F, 29.6 L/h and Vd/F(central), 3,080 liters. Tumor type (except acute myeloid leukemia), Asian race, gender, body weight, and elevated Eastern Cooperative Oncology Group score described a portion of the variability in CL/F for sunitinib and metabolite; gender and body weight explained some of the variability in Vd/F(central) for sunitinib and metabolite. Among patients, the predicted changes in sunitinib and metabolite AUC and C(max) as a result of the individual covariates ranged up to 17%. CONCLUSION: The magnitude of the predicted changes in exposure with the covariates studied minimizes the necessity for dose adjustment in any of these subpopulations.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Indoles/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Pyrroles/pharmacokinetics , Administration, Oral , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Female , Humans , Indoles/administration & dosage , Male , Middle Aged , Models, Biological , Neoplasms/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrroles/administration & dosage , Sunitinib
20.
Cancer Chemother Pharmacol ; 64(4): 691-706, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19169880

ABSTRACT

PURPOSE: The purpose of these extensive non-clinical studies was to assess pharmacokinetics and dispositional properties of sunitinib and its primary active metabolite (SU12662). METHODS: Sunitinib was administered in single and repeat oral doses in mice, rats, and monkeys. Assessments were made using liquid-chromatography-tandem mass spectrometric methods, radioactive assays, and quantitative whole body autoradiography. RESULTS: Sunitinib was readily absorbed with good oral bioavailability and linear kinetics at clinically-relevant doses. SU12662 plasma levels were less than those of sunitinib in mice and monkeys, but greater in rats. Sunitinib was extensively distributed with moderate-to-high systemic clearance and eliminated primarily into feces. Single- and repeat-dosing kinetics were similar. A prolonged half-life allowed once-daily dosing, enabling adequate systemic exposure with limited-to-moderate accumulation. In multiple-dose studies with cyclic dosing, drug plasma concentrations cleared from one cycle to the next. CONCLUSIONS: Sunitinib exhibited advantageous pharmacokinetic and dispositional properties in non-clinical species, translating into favorable properties in humans.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Indoles/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Pyrroles/pharmacokinetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/blood , Area Under Curve , Chromatography, High Pressure Liquid , Female , Indoles/administration & dosage , Indoles/blood , Macaca fascicularis , Male , Mice , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/blood , Pyrroles/administration & dosage , Pyrroles/blood , Rats , Rats, Sprague-Dawley , Sunitinib , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...