Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 17(3): 698-709, 2018 03.
Article in English | MEDLINE | ID: mdl-29444985

ABSTRACT

The development of molecularly targeted agents has benefited from use of pharmacodynamic markers to identify "biologically effective doses" (BED) below MTDs, yet this knowledge remains underutilized in selecting dosage regimens and in comparing the effectiveness of targeted agents within a class. We sought to establish preclinical proof-of-concept for such pharmacodynamics-based BED regimens and effectiveness comparisons using MET kinase small-molecule inhibitors. Utilizing pharmacodynamic biomarker measurements of MET signaling (tumor pY1234/1235MET/total MET ratio) in a phase 0-like preclinical setting, we developed optimal dosage regimens for several MET kinase inhibitors and compared their antitumor efficacy in a MET-amplified gastric cancer xenograft model (SNU-5). Reductions in tumor pY1234/1235MET/total MET of 95%-99% were achievable with tolerable doses of EMD1214063/MSC2156119J (tepotinib), XL184 (cabozantinib), and XL880/GSK1363089 (foretinib), but not ARQ197 (tivantinib), which did not alter the pharmacodynamic biomarker. Duration of kinase suppression and rate of kinase recovery were specific to each agent, emphasizing the importance of developing customized dosage regimens to achieve continuous suppression of the pharmacodynamic biomarker at the required level (here, ≥90% MET kinase suppression). The customized dosage regimen of each inhibitor yielded substantial and sustained tumor regression; the equivalent effectiveness of customized dosage regimens that achieve the same level of continuous molecular target control represents preclinical proof-of-concept and illustrates the importance of proper scheduling of targeted agent BEDs. Pharmacodynamics-guided biologically effective dosage regimens (PD-BEDR) potentially offer a superior alternative to pharmacokinetic guidance (e.g., drug concentrations in surrogate tissues) for developing and making head-to-head comparisons of targeted agents. Mol Cancer Ther; 17(3); 698-709. ©2018 AACR.


Subject(s)
Drug Development/methods , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Anilides/pharmacology , Animals , Cell Line, Tumor , Humans , Mice, Nude , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-met/metabolism , Pyridazines/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Quinolines/pharmacology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Treatment Outcome , Tumor Burden/drug effects
2.
Hum Gene Ther Clin Dev ; 25(1): 16-27, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24649838

ABSTRACT

Herpes simplex virus type 1 (HSV-1) mutants lacking the γ(1)34.5 neurovirulence loci are promising agents for treating malignant glioma. Arming oncolytic HSV-1 to express immunostimulatory genes may potentiate therapeutic efficacy. We have previously demonstrated improved preclinical efficacy, biodistribution, and safety of M002, a γ(1)34.5-deleted HSV-1 engineered to express murine IL-12. Herein, we describe the safety and biodistribution of M032, a γ(1)34.5-deleted HSV-1 virus that expresses human IL-12 after intracerebral administration to nonhuman primates, Aotus nancymae. Cohorts were administered vehicle, 10(6), or 10(8) pfu of M032 on day 1 and subjected to detailed clinical observations performed serially over a 92-day trial. Animals were sacrificed on days 3, 31, and 91 for detailed histopathologic assessments of all organs and to isolate and quantify virus in all organs. With the possible exception of one animal euthanized on day 16, neither adverse clinical signs nor sex- or dose-related differences were attributed to M032. Elevated white blood cell and neutrophil counts were observed in virus-injected groups on day 3, but no other significant changes were noted in clinical chemistry or coagulation parameters. Minimal to mild inflammation and fibrosis detected, primarily in meningeal tissues, in M032-injected animals on days 3 and 31 had mostly resolved by day 91. The highest viral DNA levels were detected at the injection site and motor cortex on day 3 but decreased in central nervous system tissues over time. These data demonstrate the requisite safety of intracerebral M032 administration for consideration as a therapeutic for treating malignant brain tumors.


Subject(s)
Glioma/therapy , Herpesvirus 1, Human/genetics , Infusions, Intraventricular , Interleukin-12/genetics , Oncolytic Virotherapy/methods , Animals , Aotidae , Brain Neoplasms/therapy , Drug Administration Routes , Female , Interleukin-12/biosynthesis , Male , Virus Replication
3.
J Virol ; 86(5): 2750-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22171271

ABSTRACT

A dependence of poliovirus on an unorthodox translation initiation mode can be targeted selectively to drive viral protein synthesis and cytotoxicity in malignant cells. Transformed cells are naturally susceptible to poliovirus, due to widespread ectopic upregulation of the poliovirus receptor, Necl-5, in ectodermal/neuroectodermal cancers. Viral tumor cell killing and the host immunologic response it engenders produce potent, lasting antineoplastic effects in animal tumor models. Clinical application of this principle depends on unequivocal demonstration of safety in primate models for paralytic poliomyelitis. We conducted extensive dose-range-finding, toxicity, biodistribution, shedding, and neutralizing antibody studies of the prototype oncolytic poliovirus recombinant, PVS-RIPO, after intrathalamic inoculation in Macaca fascicularis. These studies suggest that intracerebral PVS-RIPO inoculation does not lead to viral propagation in the central nervous system (CNS), does not cause histopathological CNS lesions or neurological symptoms that can be attributed to the virus, is not associated with extraneural virus dissemination or replication and does not induce shedding of virus with stool. Intrathalamic PVS-RIPO inoculation induced neutralizing antibody responses against poliovirus serotype 1 in all animals studied.


Subject(s)
Chimera/physiology , Disease Models, Animal , Macaca fascicularis , Poliomyelitis/virology , Poliovirus/physiology , Poliovirus/pathogenicity , Rhinovirus/genetics , Virus Shedding , Animals , Antibodies, Viral/immunology , Cell Line , Chimera/genetics , Humans , Macaca fascicularis/immunology , Macaca fascicularis/virology , Poliomyelitis/immunology , Poliovirus/genetics , Rhinovirus/physiology , Tissue Distribution , Virulence
4.
Toxicol Pathol ; 39(4): 614-22, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21558467

ABSTRACT

ST-20 (sodium 2,2-dimethylbutyrate) is a potential therapeutic agent for treatment of ß-thalassemia and sickle cell disease. A subchronic oral toxicity study was conducted in Sprague-Dawley rats (10/sex/dose) at gavage dosages of 0 (vehicle control), 200, 600, or 1,000 mg/kg, once daily for up to 15 days followed by a 14-day recovery. Ataxia (females), rough coat/thin appearance (males), and decreased body weights were observed at 1,000 mg/kg. Functional observational battery (FOB) deficits were observed more frequently in females and included decreased body tone, rectal temperature, emotional reactivity, neuromotor-neuromuscular activity (as exhibited by a deficit in visual/tactile placing accuracy, ataxia, hind limb dragging, and decreased grip strength), and rearing. ST-20 caused a decrease in WBC/RBC counts and RBC parameters; increase in reticulocytes and red cell inclusion bodies; decrease in total protein, globulin, and glucose; and increase in AG ratio. Micronucleated polychromatic erythrocytes of the bone marrow increased significantly in males at 1,000 mg/kg. Mean liver and kidney weights increased, and hepatocellular hypertrophy was observed in males at 1,000 mg/kg. Toxicologic findings were fully recovered during the 14-day recovery period. In conclusion, the no-observed adverse effect level for FOB and general toxicity was 200 mg/kg following gavage administration of ST-20 for up to 15 consecutive days.


Subject(s)
Butyrates/toxicity , Kidney/drug effects , Liver/drug effects , Organ Size/drug effects , Reticulocytes/drug effects , Toxicity Tests, Acute/methods , Administration, Oral , Animals , Body Weight , Butyrates/pharmacology , Dose-Response Relationship, Drug , Erythrocyte Count , Female , Leukocyte Count , Linear Models , Male , Micronucleus Tests , No-Observed-Adverse-Effect Level , Rats , Rats, Sprague-Dawley
5.
Hum Gene Ther ; 22(7): 821-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21171861

ABSTRACT

Conditionally replicative adenoviral (CRAd) virotherapy represents a promising therapeutic approach for cancer. We have demonstrated that a serotype chimeric adenoviral 5/3 fiber-knob modification achieves enhanced ovarian cancer infectivity, conditional replication, and oncolytic activity. This study evaluated the safety of intraperitoneal (IP) Ad5/3-Δ24 in advance of a phase I clinical trial in gynecologic cancers. Syrian hamster cohorts were treated with IP Ad5/3-Δ24 or control buffer for 3 consecutive days and euthanized on study days 8, 17, 57, and 89. Blood and tissue samples were harvested from each animal. For biodistribution studies, presence and quantitation of viral levels within samples were determined via quantitative polymerase chain reaction. For safety studies, animals were assessed for adverse vector-related tissue or laboratory effects. In the biodistribution study, low levels of Ad5/3-Δ24 DNA were noted outside of the abdominal cavity. Viral DNA levels in tissues obtained from the peritoneal cavity peaked at day 8 and declined thereafter. In the safety study, no specific histopathologic changes were attributable to virus administration. Hematologic findings noted in the 1 × 10(11) viral particles (vp)/dose group on Days 4 and/or 8 were indicative of an Ad5/3-Δ24-specific generalized inflammatory response; these findings resolved by day 56. The no observable adverse effect level was determined to be 1 × 10(10) vp/dose. This study elucidates the safety profile of IP administration of the serotype chimeric infectivity-enhanced CRAd, Ad5/3-Δ24, and provides guidance for a planned phase I trial for patients with recurrent gynecologic cancers.


Subject(s)
Adenoviridae/genetics , DNA, Viral/genetics , Oncolytic Virotherapy/methods , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/virology , Adenoviridae/physiology , Animals , Antibodies, Neutralizing/blood , Cricetinae , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Genetic Therapy , Genetic Vectors/pharmacology , Injections, Intraperitoneal , Mesocricetus , Polymerase Chain Reaction , Serotyping , Tissue Distribution , Virus Replication
6.
Cancer Chemother Pharmacol ; 63(6): 983-95, 2009 May.
Article in English | MEDLINE | ID: mdl-18766340

ABSTRACT

BACKGROUND: STAT3 overexpression has been detected in several cancers including head and neck squamous cell carcinoma (HNSCC). Previous studies using intratumoral administration of a STAT3 decoy oligonucleotide that abrogates STAT3-mediated gene transcription in preclinical cancer models have demonstrated antitumor efficacy. This study was conducted to observe the toxicity and biologic effects of the STAT3 decoy in a non-human primate model, in anticipation of initiating a clinical trial in HNSCC patients. METHODS: Three study groups (two monkeys/sex/group) were administered a single intramuscular injection of low dose of STAT3 decoy (0.8 mg total dose/monkey), high dose of STAT3 decoy (3.2 mg total dose/monkey) or vehicle control (PBS alone) on day 1 and necropsies were performed on days 2 and 15 (one monkey/sex/group/day). Low and high doses of the decoy were administered in the muscle in a volume of 0.9 ml. Tissue and blood were harvested for toxicology and biologic analyses. RESULTS: Upon observation, the STAT3 decoy-treated animals exhibited behavior that was similar to the vehicle control group. Individual animal body weights remained within 1% of pretreatment weights throughout the study. Hematological parameters were not significantly different between the control and the treatment groups. Clinical chemistry fluctuations were considered within normal limits and were not attributed to the STAT3 decoy. Assessment of complement activation breakdown product (Bb) levels demonstrated no activation of the alternative pathway of complement in any animal at any dose level. At necropsy, there were no gross or microscopic findings attributed to STAT3 decoy in any organ examined. STAT3 target gene expression at the injection site revealed decreased Bcl-X(L) and cyclin D1 expression levels in the animals treated with high dose of STAT3 decoy compared to the animals injected with low dose of STAT3 decoy or the vehicle as control. CONCLUSION: Based on these findings, the no-observable-adverse-effect-level (NOAEL) was greater than 3.2 mg/kg when administered as a single dose to male and female Cynomolgus monkeys. Plans are underway to test the safety and biologic effects of intratumoral administration of the STAT3 decoy in HNSCC patients.


Subject(s)
Gene Expression/drug effects , Oligonucleotides/toxicity , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Cyclin D1/genetics , Female , Injections, Intramuscular , Macaca fascicularis , Male , No-Observed-Adverse-Effect Level , Oligonucleotides/administration & dosage , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/physiology , Toxicity Tests , Tubulin/genetics , bcl-X Protein/genetics
7.
Article in English | MEDLINE | ID: mdl-15905139

ABSTRACT

Methods for the measurement of gentamicin concentration in several bovine tissues were developed and validated. A novel liquid chromatographic (LC) technique employed trifluoroacetic acid in the mobile phase so that all gentamicin components co-eluted. Analytes were ionized by positive-ion pneumatically assisted electrospray and detected by selected reaction monitoring (SRM) with an LC-tandem mass spectrometer (LC/MS/MS). Calibration of plasma and urine samples was based on tobramycin internal standard. Calibration of milk and kidney samples was based on external standard, due to variability of tobramycin response in these matrices. The extraction technique employed treatment with aqueous trichloroacetic acid to both precipitate protein and liberate gentamicin from the matrix. Milk samples had to be defatted by centrifugation prior to extraction. Urine samples were further cleaned up with C-18 solid phase extraction (SPE). These methods were validated for use in several residue depletion studies (reported elsewhere) to monitor the depletion of gentamicin in tissues under various dosing conditions. The plasma method was calibrated from 1 to 5000 ng/mL in two ranges, with a limit of quantitation (LOQ) in the low range calculated at 3.3 ng/mL. The milk method was calibrated from 2.5 to 2500 ng/mL with an LOQ calculated at 4.5 ng/mL. The urine method was designed for use at low levels, and was calibrated from 1 to 100 ng/mL with an LOQ of 3.8 ng/mL. The kidney method was primarily designed for analysis of small samples (approximately 100mg). This method was calibrated from 10 to 50,000 ng/g with an LOQ of 26 ng/g.


Subject(s)
Chromatography, Liquid/methods , Gentamicins/analysis , Gentamicins/metabolism , Kidney/chemistry , Mass Spectrometry/methods , Milk/chemistry , Animals , Biopsy/veterinary , Cattle , Chemical Fractionation/methods , Female
8.
Article in English | MEDLINE | ID: mdl-15358305

ABSTRACT

An LC-MS/MS method was developed to quantitate the potential antitumor agent halofuginone in plasma. The assay uses 0.2 ml of plasma; chlorohalofuginone internal standard; acetonitrile for protein precipitation; a Phenomenex SYNERGI 4 micro Polar RP 80A (4 microm, 100 mm x 2 mm) column; an isocratic mobile phase of methanol:water:formic acid (80:20:0.02, v/v/v); and positive-ion electrospray ionization with selective reaction monitoring detection. Halofuginone eluted at approximately 2.4 min, internal standard eluted at approximately 2.9 min, and no endogenous materials interfered with their measurement. The assay was accurate, precise, and linear between 0.1 and 100 ng/ml. Halofuginone could be quantitated in dog plasma for at least 24 h after an i.v. dose of 0.1mg/kg. The assay is being used in ongoing pharmacokinetic studies of halofuginone.


Subject(s)
Coccidiostats/blood , Quinazolines/blood , Animals , Chromatography, Liquid , Coccidiostats/pharmacokinetics , Dogs , Indicators and Reagents , Mass Spectrometry , Molecular Weight , Piperidines , Quinazolines/pharmacokinetics , Quinazolinones , Reference Standards , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization
9.
J Pharm Biomed Anal ; 35(1): 143-53, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15030889

ABSTRACT

A rapid and simple procedure for determination of enrofloxacin and ciprofloxacin in bovine milk and plasma is described. Protein precipitation from both milk and plasma samples was achieved by addition of acetonitrile and phosphoric acid. Acetonitrile was removed with methylene chloride, leaving enrofloxacin and ciprofloxacin in the acidic aqueous extract. The aqueous extract was analyzed by high-performance liquid chromatography (HPLC) with fluorescence detection. The limit of quantitation (LOQ) for enrofloxacin and ciprofloxacin in milk was found to be 2ng/ml. LOQ for enrofloxacin and ciprofloxacin in plasma was found to be 1ng/ml. Linear calibration curves were obtained with correlation coefficient (r(2)) >/=0.99. Analysis of quality control (QC) samples gave results within +/-10% of the nominal values. Inter-assay precision for the analysis of milk QC samples were in the ranges: 4.63-12.49% (for enrofloxacin) and 4.67-9.86% (for ciprofloxacin). Inter-assay precision for the analysis of plasma QC samples were in the ranges: 6.60-17.31% (for enrofloxacin) and 6.14-13.87% (for ciprofloxacin). Intra-assay precision for the analysis of milk QC samples were in the following ranges: 3.65-7.21% (for enrofloxacin) and 1.58-14.28% (for ciprofloxacin). Intra-assay precision for the analysis of plasma QC samples were in the following ranges: 2.17-16.95% (for enrofloxacin) and 3.31-16.31% (for ciprofloxacin). The effectiveness of protein precipitants other than phosphoric acid was investigated. The method described has been applied to a study of the pharmacokinetics of enrofloxacin and ciprofloxacin in lactating dairy cows and beef steers.


Subject(s)
Ciprofloxacin/analysis , Fluoroquinolones/analysis , Milk/chemistry , Quinolones/analysis , Animals , Calibration , Cattle , Chromatography, High Pressure Liquid/methods , Ciprofloxacin/blood , Enrofloxacin , Fluoroquinolones/blood , Molecular Structure , Quinolones/blood , Reference Standards , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Fluorescence/methods
10.
Antimicrob Agents Chemother ; 48(3): 954-60, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14982789

ABSTRACT

Artemisinin and its derivatives, artesunate and artemether, are rapidly acting antimalarials that are used for the treatment of severe and uncomplicated multidrug-resistant falciparum malaria. To optimize treatment regimens that use this new class of antimalarials, there is a need for readily available and reproducible assays to monitor drug levels closely in patients. A sensitive and reproducible bioassay for the measurement of the concentrations of artemisinin derivatives in plasma and serum is described. By modifying the in vitro drug susceptibility test, it was found that antimalarial activity in plasma or serum containing an unknown concentration of drug could be equated to the known concentrations of dihydroartemisinin (DHA) required to inhibit parasite growth. Dose-response curves for a Plasmodium falciparum clone (clone W2) and DHA were used as a standard for each assay. Assays with plasma or serum spiked with DHA proved to be reproducible (coefficient of variation,

Subject(s)
Antimalarials/blood , Artemisinins/blood , Plasmodium falciparum/drug effects , Sesquiterpenes/blood , Animals , Antimalarials/pharmacology , Artemisinins/pharmacology , Artesunate , Biological Assay , Biotransformation , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Electrochemistry , Hot Temperature , Humans , In Vitro Techniques , Macaca mulatta , Malaria, Falciparum/blood , Reproducibility of Results , Sesquiterpenes/pharmacology
11.
Toxicol Appl Pharmacol ; 187(3): 198-208, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12662903

ABSTRACT

The pregnant Holstein cow and her newborn calf were evaluated as an animal model to study in utero and for lactational drug transfer and offspring exposure. A nonsteroidal antiinflammatory drug, phenylbutazone, and an antiparasitic drug, ivermectin, were tested in the model. Prior to parturition, pregnant cows were dosed orally to steady state with phenylbutazone at 4 g/day or given a single subcutaneous injection of 200 microg ivermectin/kg body wt. The level of drug transferred to calves exposed in utero, in utero combined with lactational exposure, and via lactational exposure only, was measured from days 1 through 7 postpartum. At birth the plasma level in phenylbutazone-exposed calves was approximately one-half the dam's steady-state level. For ivermectin-exposed calves, plasma levels were at or below the limit of quantitation (0.5 ng/ml) at birth, suggesting that placental transfer of ivermectin is limited in the cow. For both drugs, rapid accumulation of the drug in calf plasma occurred with lactational exposure to a mean daily dose of 2 microg ivermectin/kg body wt or 0.1 mg phenylbutazone/kg body wt/day for the first 7 days of life. The accumulation observed in the newborn calf is attributed to the lipid solubility and long elimination half-lives of these drugs. These results demonstrate that drug transfer and offspring exposure can be studied using the cow-calf model. The data also highlight the importance of considering not only the dose but also physicochemical characteristics and pharmacokinetics of the drug in the offspring when evaluating the safety of a newborn's exposure to a drug in breast milk.


Subject(s)
Ivermectin/blood , Ivermectin/pharmacokinetics , Maternal-Fetal Exchange , Milk/chemistry , Phenylbutazone/blood , Phenylbutazone/pharmacokinetics , Animals , Animals, Newborn/blood , Animals, Suckling/blood , Anthelmintics/administration & dosage , Anthelmintics/analysis , Anthelmintics/blood , Anthelmintics/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/analysis , Anti-Inflammatory Agents, Non-Steroidal/blood , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Cattle , Female , Half-Life , Ivermectin/administration & dosage , Ivermectin/analysis , Lactation , Models, Animal , Phenylbutazone/administration & dosage , Phenylbutazone/analysis , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...