Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
ChemMedChem ; 11(23): 2621-2629, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27805767

ABSTRACT

The development of novel thalidomide derivatives as immunomodulatory and anti-angiogenic agents has revived over the last two decades. Herein we report the design and synthesis of three chemotypes of barbituric acids derived from the thalidomide structure: phthalimido-, tetrafluorophthalimido-, and tetrafluorobenzamidobarbituric acids. The latter were obtained by a new tandem reaction, including a ring opening and a decarboxylation of the fluorine-activated phthalamic acid intermediates. Thirty compounds of the three chemotypes were evaluated for their anti-angiogenic properties in an ex vivo assay by measuring the decrease in microvessel outgrowth in rat aortic ring explants. Tetrafluorination of the phthalimide moiety in tetrafluorophthalimidobarbituric acids was essential, as all of the nonfluorinated counterparts lost anti-angiogenic activity. An opening of the five-membered ring and the accompanying increased conformational freedom, in case of the corresponding tetrafluorobenzamidobarbituric acids, was well tolerated. Their activity was retained, although their molecular structures differ in torsional flexibility and possible hydrogen-bond networking, as revealed by comparative X-ray crystallographic analyses.


Subject(s)
Angiogenesis Inhibitors/chemistry , Barbiturates/chemistry , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/pharmacology , Animals , Aorta/drug effects , Aorta/physiology , Barbiturates/chemical synthesis , Barbiturates/pharmacology , Crystallography, X-Ray , Molecular Conformation , Phthalimides/chemistry , Rats , Structure-Activity Relationship , Thalidomide/chemistry
2.
Mol Cancer Ther ; 14(10): 2228-37, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26269604

ABSTRACT

Thalidomide has demonstrated clinical activity in various malignancies affecting immunomodulatory and angiogenic pathways. The development of novel thalidomide analogs with improved efficacy and decreased toxicity is an ongoing research effort. We recently designed and synthesized a new class of compounds, consisting of both tetrafluorinated thalidomide analogues (Gu973 and Gu998) and tetrafluorobenzamides (Gu1029 and Gu992). In this study, we demonstrate the antiangiogenic properties of these newly synthesized compounds. We examined the specific antiangiogenic characteristics in vitro using rat aortic rings with carboxyamidotriazole as a positive control. In addition, further in vitro efficacy was evaluated using human umbilical vein endothelial cells (HUVEC) and PC3 cells treated with 5 and 10 µmol/L doses of each compound. All compounds were seen to reduce microvessel outgrowth in rat aortic rings as well as to inhibit HUVECs to a greater extent, at lower concentrations than previously tested thalidomide analogs. The antiangiogenic properties of the compounds were also examined in vivo in fli1:EGFP zebrafish embryos, where all compounds were seen to inhibit the extent of outgrowth of newly developing blood vessels. In addition, Gu1029 and Gu973 reduced the anti-inflammatory response in mpo:GFP zebrafish embryos, whereas Gu998 and Gu992 showed no difference. The compounds' antitumor effects were also explored in vivo using the human prostate cancer PC3 xenograft model. All four compounds were also screened in vivo in chicken embryos to investigate their teratogenic potential. This study establishes these novel thalidomide analogues as a promising immunomodulatory class with anticancer effects that warrant further development to characterize their mechanisms of action.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Hydrocarbons, Fluorinated/pharmacology , Neovascularization, Pathologic/prevention & control , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Chick Embryo , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Inhibitory Concentration 50 , Male , Rats, Sprague-Dawley , Tumor Burden , Xenograft Model Antitumor Assays , Zebrafish
3.
Oncotarget ; 5(18): 8161-72, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25327558

ABSTRACT

Nelfinavir is an HIV protease inhibitor being repurposed as an anti-cancer agent in preclinical models and in small oncology trials, yet the MTD of nelfinavir has not been determined. Therefore, we conducted a Phase Ia study to establish the maximum tolerated dose (MTD) and dose limiting toxicities (DLT) of nelfinavir in subjects with advanced solid tumors. Adults with refractory cancers were given oral nelfinavir twice daily with pharmacokinetic and pharmacodynamic analyses. Twenty-eight subjects were enrolled. Nelfinavir was generally well tolerated. Common adverse events included diarrhea, anemia, and lymphopenia, which were mostly mild. The DLT was rapid-onset neutropenia that was reversible. The MTD was established at 3125 mg twice daily. In an expansion cohort at the MTD, one of 11 (9%) evaluable subjects had a confirmed partial response. This, plus two minor responses, occurred in subjects with neuroendocrine tumors of the midgut or pancreatic origin. Thirty-six percent of subjects had stable disease for more than 6 months. In peripheral blood mononuclear cells, Nelfinavir inhibited AKT and induced markers of ER stress. In summary, nelfinavir is well tolerated in cancer patients at doses 2.5 times the FDA-approved dose for HIV management and showed preliminary activity in tumors of neuroendocrine origin.


Subject(s)
Antineoplastic Agents/therapeutic use , HIV Protease Inhibitors/therapeutic use , Nelfinavir/therapeutic use , Neoplasms/drug therapy , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Drug Administration Schedule , Female , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/pharmacokinetics , Humans , Male , Maryland , Maximum Tolerated Dose , Middle Aged , Nelfinavir/administration & dosage , Nelfinavir/adverse effects , Nelfinavir/pharmacokinetics , Neoplasms/pathology , Time Factors , Tomography, X-Ray Computed , Treatment Outcome , Young Adult
4.
AAPS PharmSciTech ; 13(2): 661-73, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22552929

ABSTRACT

A stability-indicating high-performance liquid chromatography method to quantify 2-(2,4-difluorophenyl)-4,5,6,7-tetrafluoroisoindoline-1,3-dione (NSC-726796) and its three main degradation products was developed. This method was used to investigate its degradation kinetics and mechanism. The reaction follows first-order kinetics and appears to be base catalyzed with the maximum stability at pH 1. The products were identified as 2-(2,4-difluorophenylcarbamoyl)-3,4,5,6-tetrafluorobenzoic acid (NSC-749820), 2,4-difluoroaniline, and tetrafluorophthalic acid. The parent drug, NSC-726796, was also found to react with methanol and ethanol. NSC-726796 demonstrates antiangiogenic activity, however, when its degradant NSC749820 does not show antiangiogenic activity.


Subject(s)
Angiogenesis Inhibitors/chemistry , Phthalimides/chemistry , Angiogenesis Inhibitors/pharmacology , Aniline Compounds/chemistry , Animals , Calorimetry, Differential Scanning , Chemistry, Pharmaceutical , Chromatography, High Pressure Liquid , Drug Stability , Ethanol/chemistry , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Methanol/chemistry , Neovascularization, Physiologic/drug effects , Phthalic Acids/chemistry , Phthalimides/pharmacology , Rats , Reproducibility of Results , Solvents/chemistry , Technology, Pharmaceutical/methods , Temperature , Tissue Culture Techniques
5.
Clin Cancer Res ; 18(7): 2099-107, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22307138

ABSTRACT

PURPOSE: Several case reports suggest sorafenib exposure and sorafenib-induced hyperbilirubinemia may be related to a (TA)(5/6/7) repeat polymorphism in UGT1A1*28 (UGT, uridine glucuronosyl transferase). We hypothesized that sorafenib inhibits UGT1A1 and individuals carrying UGT1A1*28 and/or UGT1A9 variants experience greater sorafenib exposure and greater increase in sorafenib-induced plasma bilirubin concentration. EXPERIMENTAL DESIGN: Inhibition of UGT1A1-mediated bilirubin glucuronidation by sorafenib was assessed in vitro. UGT1A1*28 and UGT1A9*3 genotypes were ascertained with fragment analysis or direct sequencing in 120 cancer patients receiving sorafenib on five different clinical trials. Total bilirubin measurements were collected in prostate cancer patients before receiving sorafenib (n = 41) and 19 to 30 days following treatment and were compared with UGT1A1*28 genotype. RESULTS: Sorafenib exhibited mixed-mode inhibition of UGT1A1-mediated bilirubin glucuronidation (IC(50) = 18 µmol/L; K(i) = 11.7 µmol/L) in vitro. Five patients carrying UGT1A1*28/*28 (n = 4) or UGT1A9*3/*3 (n = 1) genotypes had first dose, dose-normalized areas under the sorafenib plasma concentration versus time curve (AUC) that were in the 93rd percentile, whereas three patients carrying UGT1A1*28/*28 had AUCs in the bottom quartile of all genotyped patients. The Drug Metabolizing Enzymes and Transporters genotyping platform was applied to DNA obtained from six patients, which revealed the ABCC2-24C>T genotype cosegregated with sorafenib AUC phenotype. Sorafenib exposure was related to plasma bilirubin increases in patients carrying 1 or 2 copies of UGT1A1*28 alleles (n = 12 and n = 5; R(2) = 0.38 and R(2) = 0.77; P = 0.032 and P = 0.051, respectively). UGT1A1*28 carriers showed two distinct phenotypes that could be explained by ABCC2-24C>T genotype and are more likely to experience plasma bilirubin increases following sorafenib if they had high sorafenib exposure. CONCLUSIONS: This pilot study indicates that genotype status of UGT1A1, UGT1A9, and ABCC2 and serum bilirubin concentration increases reflect abnormally high AUC in patients treated with sorafenib.


Subject(s)
Benzenesulfonates/pharmacokinetics , Glucuronosyltransferase/genetics , Pyridines/pharmacokinetics , Aged , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Benzenesulfonates/adverse effects , Benzenesulfonates/metabolism , Bilirubin/metabolism , Clinical Trials as Topic , Disease-Free Survival , Female , Genotype , Glucuronides/metabolism , Glucuronosyltransferase/antagonists & inhibitors , Glucuronosyltransferase/metabolism , Humans , Hyperbilirubinemia/chemically induced , Hyperbilirubinemia/genetics , Male , Middle Aged , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Niacinamide/analogs & derivatives , Pharmacogenetics , Phenylurea Compounds , Pilot Projects , Polymorphism, Genetic , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Pyridines/adverse effects , Pyridines/metabolism , Sorafenib , UDP-Glucuronosyltransferase 1A9
6.
Invest New Drugs ; 30(1): 90-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-20820910

ABSTRACT

LP-261 is a novel tubulin targeting anticancer agent that binds at the colchicine site on tubulin, inducing G2/M arrest. Screening in the NCI60 cancer cell lines resulted in a mean GI50 of approximately 100 nM. Here, we report the results of testing in multiple mouse xenograft models and angiogenesis assays, along with bioavailability studies. To determine the antiangiogenic activity of LP-261, both in vitro and ex vivo experiments were performed. Human umbilical vein endothelial cells (HUVECs) were incubated with LP-261 at 50 nM to 10 µM. LP-261 was also tested in a rat aortic ring assay, from 20 nM to 10 µM. Multiple mouse xenograft studies were performed to assess in vivo antitumor activity. LP-261 was tested as a single agent in colon adenocarcinoma (SW620) and prostate cancer (LNCaP and PC3) xenografts, evaluating several different dosing schedules. LP-261 was also used in combination with bevacizumab in the SW620 xenograft model. LP-261 also exhibited high oral bioavailability and apparent lack of efflux by intestinal transporters such as ABCB1. LP-261 is a very potent inhibitor of angiogenesis, preventing microvessel outgrowth in the rat aortic ring assay and HUVEC cell proliferation at nanomolar concentrations. Complete inhibition of tumor growth was achieved in the PC3 xenograft model and shown to be schedule dependent. Excellent inhibition of tumor growth in the SW620 model was observed, comparable with paclitaxel. Combining oral, low dose LP-261 with bevacizumab led to significantly improved tumor inhibition. Oral LP-261 is very effective at inhibiting tumor growth in multiple mouse xenograft models and is well tolerated.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Administration, Oral , Angiogenesis Inhibitors/administration & dosage , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/blood , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Bevacizumab , Biological Availability , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Intestinal Absorption , Intestinal Mucosa/metabolism , Isonicotinic Acids/administration & dosage , Male , Mice , Mice, Nude , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Physiologic/drug effects , Paclitaxel/administration & dosage , Permeability , Rats , Rats, Sprague-Dawley , Sulfonamides/administration & dosage , Time Factors , Tubulin Modulators/administration & dosage , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
7.
Br J Clin Pharmacol ; 72(2): 294-305, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21392074

ABSTRACT

WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT: Sorafenib is a multikinase inhibitor with activity against B-raf, C-raf, VEGFR2, PDGFRß and FGFR1. Sorafenib is clinically approved for the treatment of renal cell carcinoma (RCC) and hepatocellular carcinoma (HCC). The pharmacokinetics (PK) of sorafenib are highly variable between subjects. Sorafenib exposure increases less than dose proportionally (likely due to limited solubility). Sorafenib undergoes enterohepatic recycling (EHC). WHAT THIS STUDY ADDS: This is the first study to characterize the PK of sorafenib using a model based on sorafenib's known disposition characteristics such as delayed/solubility-limited GI absorption and EHC. The parameterization of the EHC model used a square wave function to describe the gall bladder emptying. This study evaluated the effect of baseline bodyweight, BSA, age, gender, liver function parameters, kidney function parameters and genotype with respect to CYP3A4*1B, CYP3A5*3C, UGT1A9*3 and UGT1A9*5 on sorafenib PK. No clinically important covariates were identified. This model can be used to simulate and explore alternative dosing regimens and to develop exposure-response relationships for sorafenib. AIMS: To characterize the pharmacokinetics (PK) of sorafenib in patients with solid tumours and to evaluate the possible effects of demographic, clinical and pharmacogenetic (CYP3A4*1B, CYP3A5*3C, UGT1A9*3 and UGT1A9*5) covariates on the disposition of sorafenib. METHODS: PK were assessed in 111 patients enrolled in five phase I and II clinical trials, where sorafenib 200 or 400 mg was administered twice daily as a single agent or in combination therapy. All patients were genotyped for polymorphisms in metabolic enzymes for sorafenib. Population PK analysis was performed by using nonlinear mixed effects modelling (NONMEM). The final model was validated using visual predictive checks and nonparametric bootstrap analysis. RESULTS: A one compartment model with four transit absorption compartments and enterohepatic circulation (EHC) adequately described sorafenib disposition. Baseline bodyweight was a statistically significant covariate for distributional volume, accounting for 4% of inter-individual variability (IIV). PK model parameter estimates (range) for an 80 kg patient were clearance 8.13 l h(-1) (3.6-22.3 l h(-1) ), volume 213 l (50-1000 l), mean absorption transit time 1.98 h (0.5-13 h), fraction undergoing EHC 50% and average time to gall bladder emptying 6.13 h. CONCLUSIONS: Overall, population PK analysis was consistent with known biopharmaceutical/PK characteristics of oral sorafenib. No clinically important PK covariates were identified.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Benzenesulfonates/pharmacokinetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Renal Cell/metabolism , Liver Neoplasms/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyridines/pharmacokinetics , Aged , Aged, 80 and over , Cytochrome P-450 CYP3A/metabolism , Female , Glucuronosyltransferase/metabolism , Humans , Male , Middle Aged , Models, Theoretical , Niacinamide/analogs & derivatives , Phenylurea Compounds , Protein-Tyrosine Kinases/metabolism , Sorafenib , Statistics as Topic , UDP-Glucuronosyltransferase 1A9
8.
Clin Cancer Res ; 17(4): 937-46, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21106724

ABSTRACT

PURPOSE: Although the ABCB1 (P-glycoprotein) drug transporter is a constituent of several blood-tissue barriers (i.e., blood-brain and blood-nerve), its participation in a putative blood-heart barrier has been poorly explored. ABCB1 could decrease the intracardiac concentrations of drugs that cause QT prolongation and cardiotoxicity. EXPERIMENTAL DESIGN: ABCB1-related romidepsin transport kinetics were explored in LLC-PK1 cells transfected with different ABCB1 genetic variants. ABCB1 plasma and intracardiac concentrations were determined in Abcb1a/1b (-/-) mice and wild-type FVB controls. These same mice were used to evaluate romidepsin-induced heart rate-corrected QT interval (QTc) prolongation over time. Finally, a cohort of 83 individuals with available QTcB and ABCB1 genotyping data were used to compare allelic variation in ABCB1 versus QTc-prolongation phenotype. RESULTS: Here, we show that mice lacking the ABCB1-type P-glycoprotein have higher intracardiac concentrations of a model ABCB1 substrate, romidepsin, that correspond to changes in QT prolongation from baseline (ΔQTc) over time. Consistent with this observation, we also show that patients carrying genetic variants that could raise ABCB1 expression in the cardiac endothelium have lower ΔQTc following a single dose of romidepsin. CONCLUSIONS: To our knowledge, this is the first evidence that Abcb1-type P-glycoprotein can limit intracardiac exposure to a drug that mediates QT prolongation and suggests that certain commonly inherited polymorphisms in ABCB1 may serve as markers for QT prolongation following the administration of ABCB1-substrate drugs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Arrhythmias, Cardiac/chemically induced , Myocardium/metabolism , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Line , Depsipeptides/adverse effects , Depsipeptides/pharmacokinetics , Depsipeptides/pharmacology , Female , Gene Frequency , Genetic Association Studies , Genotype , Humans , Male , Mice , Mice, Knockout , Middle Aged , Polymorphism, Single Nucleotide
9.
Clin Cancer Res ; 17(3): 569-80, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21081657

ABSTRACT

PURPOSE: P-glycoprotein (Pgp) antagonists have been difficult to develop because of complex pharmacokinetic interactions and a failure to show meaningful results. Here we report the results of a pharmacokinetic and pharmacodynamic trial using a third-generation, potent, noncompetitive inhibitor of Pgp, tariquidar (XR9576), in combination with docetaxel. EXPERIMENTAL DESIGN: In the first treatment cycle, the pharmacokinetics of docetaxel (40 mg/m(2)) were evaluated after day 1 and day 8 doses, which were administered with or without tariquidar (150 mg). (99m)Tc-sestamibi scanning and CD56(+) mononuclear cell rhodamine efflux assays were conducted to assess Pgp inhibition. In subsequent cycles, 75 mg/m(2) docetaxel was administered with 150 mg tariquidar every 3 weeks. RESULTS: Forty-eight patients were enrolled onto the trial. Nonhematologic grade 3/4 toxicities in 235 cycles were minimal. Tariquidar inhibited Pgp-mediated rhodamine efflux from CD56(+) cells and reduced (99m)Tc-sestamibi clearance from the liver. There was striking variability in basal sestamibi uptake; a 12% to 24% increase in visible lesions was noted in 8 of 10 patients with lung cancer. No significant difference in docetaxel disposition was observed in pairwise comparison with and without tariquidar. Four partial responses (PR) were seen (4/48); 3 in the non-small cell lung cancer (NSCLC) cohort, measuring 40%, 57%, and 67% by RECIST, and 1 PR in a patient with ovarian cancer. CONCLUSIONS: Tariquidar is well tolerated, with less observed systemic pharmacokinetic interaction than previous Pgp antagonists. Variable effects of tariquidar on retention of sestamibi in imageable lung cancers suggest that follow-up studies assessing tumor drug uptake in this patient population would be worthwhile.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lung Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Quinolines/administration & dosage , Taxoids/administration & dosage , Uterine Cervical Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Adolescent , Adult , Aged , Aged, 80 and over , Docetaxel , Female , Humans , Middle Aged , Quinolines/pharmacokinetics
10.
J Chromatogr B Analyt Technol Biomed Life Sci ; 878(30): 3187-92, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20951100

ABSTRACT

A simple, rapid and sensitive liquid chromatography/tandem mass spectrometric (LC/MS/MS) analytical method was developed for quantification of Hsp90 inhibitor PF-04928473 in human plasma, following administration of its prodrug, PF-04929113. Sample processing involved protein precipitation by addition of 0.4 mL of methanol containing internal standard (PF-04972487) to 50 µL volume of plasma sample. Chromatographic separation of PF-04928473 and PF-04972487 was achieved on a Phenomenex®) Luna C18(2) (2.0 mm x 50 mm, 5 µm) column using a gradient elution method with mobile phase solvents: methanol containing 0.1% formic acid and 0.1% formic acid at a flow rate of 0.25 mL/min. Detection was performed in electrospray positive ionization mode, monitoring the ion transitions from m/z 465.1→350.1 (PF-04928473) and m/z 447.0→329.1 (PF-04972487). The retention times for PF-04928473 and PF-04972487 were 1.86 and 2.85 min, respectively. Calibration curves were generated in the range of 2-2000 ng/mL. The accuracy and precision ranged from 94.1 to 99.0% and 86.7 to 97.6%, respectively, which were calculated using quality control samples of three different concentrations analyzed in quintuplicate on four different days.


Subject(s)
Chromatography, High Pressure Liquid/methods , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Heterocyclic Compounds, 4 or More Rings/blood , Tandem Mass Spectrometry/methods , Humans , Spectrometry, Mass, Electrospray Ionization
11.
Expert Rev Anticancer Ther ; 10(7): 997-1008, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20645688

ABSTRACT

Romidepsin is a histone deacetylase inhibitor (HDI), approved by the US FDA for the treatment of cutaneous T-cell lymphoma (CTCL). Although various mechanisms have been proposed for the activity of HDIs, including induction of genes controlling cell cycle, acetylation of cytoplasmic proteins and direct induction of apoptosis, the mechanism underlying activity of romidepsin and other HDIs in CTCL is not known. Romidepsin induces long-lasting responses. The side-effect profile is similar to that of other HDIs, causing fatigue, nausea and thrombocytopenia. Management of the CTCL population requires vigilence to prevent infection with skin contaminants, and monitoring of potassium and magnesium, electrolytes found to be low in a large proportion of patients. Electrocardiographic (ECG) changes are common but are not associated with myocardial damage. When molecular end points were evaluated in 61 patients enrolled on a Phase II trial with romidepsin, response was associated with persistence of acetylated histone H3, suggesting that drug exposure is important in effective therapy with romidepsin. Future studies will endeavor to identify combination strategies to increase the efficacy both in resistant CTCL and in solid tumors and to identify biomarkers of response that will allow selection of patients most likely to benefit from the therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Depsipeptides/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Lymphoma, T-Cell, Cutaneous/drug therapy , Neoplasms/drug therapy , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Bone Marrow Diseases/chemically induced , Clinical Trials, Phase II as Topic , Depsipeptides/adverse effects , Depsipeptides/pharmacology , Fatigue/chemically induced , Gastrointestinal Diseases/chemically induced , Gene Targeting , Heart Diseases/chemically induced , Histone Deacetylase Inhibitors/adverse effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/physiology , Humans , Infections/etiology , Lymphoma, T-Cell, Cutaneous/enzymology , Lymphoma, T-Cell, Cutaneous/pathology , Molecular Structure , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Neoplasms/enzymology , Practice Guidelines as Topic
12.
Cancer Biol Ther ; 10(1): 38-43, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20495381

ABSTRACT

Focal adhesion kinase (FAK) is essential in regulating integrin signaling pathways responsible for cell survival and proliferation, as well as motility, making FAK a distinctive target in the field of anticancer drug development, especially with regards to metastatic disease.(1) Our objective was to demonstrate tumor growth inhibition by PF-562,271, a selective inhibitor of FAK and FAK2, or Pyk2,(2) in mouse xenograft models, both subcutaneous and metastatic, employing the human prostate cancer cell line PC3M-luc-C6, a modified PC3M cell line that expresses luciferase. After 2 weeks of treatment with PF-562,271, 25 mg/kg PO BID 5x/wk, the subcutaneous model showed a 62% tumor growth inhibition compared to control based on tumor measurements (p < 0.05), with a 88% vs. a 490% increase in bioluminescent signal for treatment and control respectively (p < 0.05). In the metastasis model, the percent change from baseline, after 18 days of treatment, of the treatment group was 2,854 vs. 14,190% for the vehicle (p < 0.01). These results show that PF-562,271 has a potent effect on metastatic prostate cancer growth in vivo.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Indoles/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays , Animals , Humans , Injections, Subcutaneous , Luminescent Measurements , Lymphatic Metastasis , Male , Mice , Mice, Nude , Phosphorylation/drug effects , Prostatic Neoplasms/enzymology , Survival Rate
13.
Mol Biotechnol ; 44(2): 152-67, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19950006

ABSTRACT

This review provides an overview of the pharmacogenetics of membrane transporters including selected ABC transporters (ABCB1, ABCC1, ABCC2, and ABCG2) and OATPs (OATP1B1 and OATP1B3). Membrane transporters are heavily involved in drug clearance and alters drug disposition by actively transporting substrate drugs between organs and tissues. As such, polymorphisms in the genes encoding these proteins may have significant effects on the absorption, distribution, metabolism and excretion of compounds, and may alter pharmacodynamics of many agents. This review discusses the techniques used to identify substrates and inhibitors of these proteins and subsequently to assess the effect of genetic mutation on transport, both in vitro and in vivo. A comprehensive list of substrates for the major drug transporters is included. Finally, studies linking transporter genotype with clinical outcomes are discussed.


Subject(s)
Membrane Transport Proteins/physiology , Pharmacogenetics/methods , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , ATP-Binding Cassette Transporters/physiology , Animals , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Protein 2 , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Organic Anion Transporters/physiology
14.
Br J Haematol ; 148(2): 256-67, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19874311

ABSTRACT

Romidepsin has shown promise in the treatment of T-cell lymphomas, and so we evaluated molecular endpoints gathered from 61 patients enrolled on a phase II trial of romidepsin in cutaneous and peripheral T-cell lymphoma at the National Institutes of Health. The endpoints included histone H3 acetylation and ABCB1 gene expression in peripheral blood mononuclear cells (PBMCs); ABCB1 gene expression in tumour biopsy samples; and blood fetal haemoglobin levels (HbF), all of which were increased following romidepsin treatment. The fold increase in histone acetylation in PBMCs at 24 h was weakly to moderately well correlated with the pharmacokinetic parameters C(max) and area under the curve (AUC)(last) (rho = 0.37, P = 0.03 and rho = 0.36, P = 0.03 respectively) and inversely associated with clearance (rho = -0.44; P = 0.03). Histone acetylation in PBMCs at 24 h was associated with response (P = 0.026) as was the increase in fetal haemoglobin (P = 0.014); this latter association may be due to the longer on-study duration for patients with disease response. Together, these results suggest that pharmacokinetics may be an important determinant of response to histone deacetylase inhibitors (HDIs) - the association with histone acetylation in PBMCs at 24 h is consistent with a hypothesis that potent HDIs are needed for a critical threshold of drug exposure and durable activity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antibiotics, Antineoplastic/therapeutic use , Depsipeptides/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Lymphoma, T-Cell, Peripheral/drug therapy , Lymphoma, T-Cell, Peripheral/metabolism , Lymphoma, T-Cell/drug therapy , Skin Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Acetylation , Biopsy , Fetal Hemoglobin/analysis , Histones/metabolism , Humans , Immunoblotting , Leukocytes, Mononuclear/metabolism , Lymphoma, T-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/metabolism
15.
Pharmacotherapy ; 30(1): 52-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20030473

ABSTRACT

STUDY OBJECTIVE: To determine if excretion of sorafenib in sweat is associated with hand-foot skin reaction in patients receiving sorafenib. DESIGN: Prospective pilot study. SETTING: Outpatient clinic of a cancer research institution. PATIENTS: Two patients who were receiving sorafenib and developed a hand-foot skin reaction of at least grade 1 and two healthy subjects (controls). INTERVENTION: Sweat production was stimulated in both the patients with hand-foot skin reaction and the healthy subjects by means of pilocarpine iontophoresis. MEASUREMENTS AND MAIN RESULTS: Sweat samples were collected from the patients with hand-foot skin reaction and from the healthy subjects. Using liquid chromatography-tandem mass spectrometry, sorafenib concentrations were measured in the sweat samples. Sweat samples from the healthy subjects were spiked with known concentrations of sorafenib to determine the lower limit of quantification of the assay, which was determined to be 5 ng/ml. Sorafenib concentrations in the samples from the patients with hand-foot skin reaction were undetectable based on the assay's sensitivity. CONCLUSION: Our results suggest that hand-foot skin reaction in patients receiving sorafenib is not associated with excretion of sorafenib in sweat. Further studies are needed to understand the mechanism of hand-foot skin reaction, a treatment-limiting adverse effect of multikinase inhibitors.


Subject(s)
Antineoplastic Agents/adverse effects , Benzenesulfonates/adverse effects , Benzenesulfonates/analysis , Drug Eruptions/etiology , Foot Dermatoses/chemically induced , Hand Dermatoses/chemically induced , Protein Kinase Inhibitors/adverse effects , Pyridines/adverse effects , Pyridines/analysis , Sweat/chemistry , Antineoplastic Agents/analysis , Antineoplastic Agents/blood , Antineoplastic Agents/metabolism , Benzenesulfonates/blood , Chromatography, High Pressure Liquid , Female , Humans , Iontophoresis , Limit of Detection , Male , Middle Aged , Niacinamide/analogs & derivatives , Phenylurea Compounds , Pilocarpine/pharmacology , Pilot Projects , Protein Kinase Inhibitors/analysis , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/metabolism , Pyridines/blood , Severity of Illness Index , Sorafenib , Tandem Mass Spectrometry
16.
Cancer Chemother Pharmacol ; 65(2): 383-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19894051

ABSTRACT

PURPOSE: UCN-01 potently inhibits protein kinase C, phosphatidylinositide-dependent kinase-1, and checkpoint kinase 1, which are involved in regulating cell cycle progression. We designed a phase I study to determine the maximum tolerated dose (MTD) of UCN-01 with prednisone in patients with advanced malignancies. METHODS: UCN-01 was administered as a continuous intravenous infusion over 72 h in cycle 1 and 36 h in subsequent cycles. Prednisone was given orally at 60 mg/m(2) per day for five consecutive days within each 28-day cycle. Standard dose escalation was employed, and MTD was defined as the dose at which no more than one of six patients experienced a dose-limiting toxicity (DLT). Plasma pharmacokinetics of UCN-01 were assessed. RESULTS: Fifteen patients received a total of 55 courses of treatment. The MTD and the recommended phase II dose of UCN-01 in this combination is 72 mg/m(2) total dose over 72 h for cycle 1 followed by 36 mg/m(2) per cycle over 36 h. All patients experienced hyperglycemia but responded to insulin treatment. Hypophosphatemia was a DLT in two patients. There were no cumulative toxicities. No objective responses were observed, but five patients had stable disease, including two patients with lymphoid malignancies who had prolonged disease stabilizations. UCN-01 has a long terminal half-life and low clearance; there was wide inter-patient variability in peak concentrations. CONCLUSION: UCN-01 can be safely administered in combination with prednisone without unacceptable toxicity. The prolonged stable disease in two patients with lymphoid malignancies is a proof of principle for the evaluation of cyclin-dependent kinase inhibitors in oncology.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Humans , Lymphoma/drug therapy , Lymphoma/pathology , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Metastasis , Neoplasms/pathology , Prednisone/administration & dosage , Staurosporine/administration & dosage , Staurosporine/analogs & derivatives , Staurosporine/pharmacokinetics , Young Adult
17.
Eur J Cancer ; 46(2): 340-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19945858

ABSTRACT

PURPOSE: Phase I dose-escalation study to determine the toxicity and maximum tolerated dose (MTD) of 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG), a heat shock protein 90 (Hsp90) inhibitor, administered on a twice weekly schedule in patients with advanced cancer. EXPERIMENTAL DESIGN: 17-DMAG was administered as a 1- to 2-h infusion twice weekly in 4-week cycles. An accelerated titration design was followed until toxicity was observed, at which point standard dose-escalation proceeded. MTD was defined as the dose at which no more than one of the six patients experienced a dose-limiting toxicity (DLT). Pharmacokinetics were assessed, and Hsp70 mRNA, whose gene product is a chaperone previously shown to be upregulated following the inhibition of Hsp90, was measured in peripheral blood mononuclear cells (PBMCs). RESULTS: A total of 31 patients received 92 courses of treatment. The MTD was 21mg/m(2)/d; 20 patients were enrolled at this dose level. Nine patients had stable disease for a median of 4 (range 2-22) months. Both C(max) and AUC increased proportionally with dose. The most common toxicities were grade 1 or 2 fatigue, anorexia, nausea, blurred vision and musculoskeletal pain. DLTs were peripheral neuropathy and renal dysfunction. Expression of Hsp70 mRNA in PBMCs was highly variable. CONCLUSION: Twice-weekly i.v. infusion of 17-DMAG is well tolerated, and combination phase I studies are warranted.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzoquinones/administration & dosage , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/administration & dosage , Neoplasms/drug therapy , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Benzoquinones/adverse effects , Benzoquinones/pharmacokinetics , Drug Administration Schedule , Female , HSP70 Heat-Shock Proteins/metabolism , Humans , Infusions, Intravenous , Lactams, Macrocyclic/adverse effects , Lactams, Macrocyclic/pharmacokinetics , Leukocytes, Mononuclear/metabolism , Male , Maximum Tolerated Dose , Middle Aged , Young Adult
18.
J Clin Oncol ; 27(32): 5410-7, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19826128

ABSTRACT

PURPOSE Romidepsin (depsipeptide or FK228) is a member of a new class of antineoplastic agents active in T-cell lymphoma, the histone deacetylase inhibitors. On the basis of observed responses in a phase I trial, a phase II trial of romidepsin in patients with T-cell lymphoma was initiated. PATIENTS AND METHODS The initial cohort was limited to patients with cutaneous T-cell lymphoma (CTCL), or subtypes mycosis fungoides or Sézary syndrome, who had received no more than two prior cytotoxic regimens. There were no limits on other types of therapy. Subsequently, the protocol was expanded to enroll patients who had received more than two prior cytotoxic regimens. Results Twenty-seven patients were enrolled onto the first cohort, and a total of 71 patients are included in this analysis. These patients had undergone a median of four prior treatments, and 62 patients (87%) had advanced-stage disease (stage IIB, n = 15; stage III, n= 6; or stage IV, n = 41). Toxicities included nausea, vomiting, fatigue, and transient thrombocytopenia and granulocytopenia. Pharmacokinetics were evaluated with the first administration of romidepsin. Complete responses were observed in four patients, and partial responses were observed in 20 patients for an overall response rate of 34% (95% CI, 23% to 46%). The median duration of response was 13.7 months. CONCLUSION The histone deacetylase inhibitor romidepsin has single-agent clinical activity with significant and durable responses in patients with CTCL.


Subject(s)
Depsipeptides/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Lymphoma, T-Cell, Cutaneous/drug therapy , Adult , Aged , Aged, 80 and over , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/therapeutic use , Area Under Curve , Depsipeptides/adverse effects , Depsipeptides/pharmacokinetics , Fatigue/chemically induced , Female , Histone Deacetylase Inhibitors/adverse effects , Histone Deacetylase Inhibitors/pharmacokinetics , Humans , Leukemia/chemically induced , Lymphoma, T-Cell, Cutaneous/pathology , Male , Metabolic Clearance Rate , Middle Aged , Nausea/chemically induced , Thrombocytopenia/chemically induced , Treatment Outcome
19.
Anticancer Agents Med Chem ; 9(10): 1058-69, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19719457

ABSTRACT

Thalidomide is emerging as a potentially important therapeutic option in the treatment of metastatic prostate cancer. Although the mechanism of action of this agent remains elusive in malignancies of the prostate, recent data has indicated that thalidomide may play a role in inflammation, immunomodulation, and anti-angiogenesis. Lenalidomide (CC-5013), a thalidomide analogue with improved activity and safety profile in certain disease contexts, is in the early stages of development in prostate cancer. This review will provide the current status of the history, mechanism, metabolism, and clinical use of thalidomide in metastatic prostate cancer. It will also describe the mechanism and clinical use of lenalidomide as it pertains to malignancies of the prostate.


Subject(s)
Antineoplastic Agents/therapeutic use , Prostatic Neoplasms/drug therapy , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Animals , Antineoplastic Agents/chemistry , Humans , Lenalidomide , Male , Prostatic Neoplasms/metabolism , Thalidomide/chemistry , Treatment Outcome
20.
J Exp Clin Cancer Res ; 28: 99, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19591692

ABSTRACT

BACKGROUND: Imatinib, a tyrosine kinase inhibitor currently approved for treatment of several malignancies, has been shown to be a substrate for multiple efflux-transporter proteins, including ABCB1 (P-glycoprotein) and ABCG2 (BCRP). The effect of inhibiting these transporters on tissue exposure to imatinib remains unclear. OBJECTIVE: To assess the role of these transporters on drug disposition, 50 mg/kg imatinib was administered to Balb/C mice, 30 minutes after receiving tariquidar (10 mg/kg), an inhibitor of both ABCB1 and ABCG2, or vehicle, via oral gavage. METHODS: Quantitative determination of imatinib in mouse plasma, liver and brain was performed using a newly-developed and validated liquid-chromatography-mass spectrometric method. RESULTS: Exposure to imatinib was 2.2-fold higher in plasma, liver and brain in mice that received tariquidar, as compared to those that received the vehicle (P = 0.001). The peak plasma concentration did not increase substantially, suggesting that tariquidar is affecting the distribution, metabolism and/or excretion of imatinib, rather than absorption. Though tariquidar increased the absolute exposure of imatinib, the brain-to-plasma ratio of imatinib was unaffected. CONCLUSION: This study suggests that intentional inhibition of ABCB1 and ABCG2 function at the blood-brain barrier is unlikely to significantly improve clinical outcome of imatinib with currently used dosing regimens.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Antineoplastic Agents/pharmacokinetics , Piperazines/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , Quinolines/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Benzamides , Imatinib Mesylate , Male , Mice , Mice, Inbred BALB C , Piperazines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...