Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
bioRxiv ; 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38405696

ABSTRACT

Victims of a radiation terrorist event will include pregnant women and unborn fetuses. Mitochondrial dysfunction and oxidative stress are key pathogenic factors of fetal irradiation injury. The goal of this preclinical study is to investigate the efficacy of mitigating fetal irradiation injury by maternal administration of the mitochondrial-targeted gramicidin S (GS)- nitroxide radiation mitigator, JP4-039. Pregnant female C57BL/6NTac mice received 3 Gy total body ionizing irradiation (TBI) at mid-gestation embryonic day 13.5 (E13.5). Using novel time- and-motion-resolved 4D in utero magnetic resonance imaging (4D-uMRI), we found TBI caused extensive injury to the fetal brain that included cerebral hemorrhage, loss of cerebral tissue, and hydrocephalus with excessive accumulation of cerebrospinal fluid (CSF). Histopathology of the fetal mouse brain showed broken cerebral vessels and elevated apoptosis. Further use of novel 4D Oxy-wavelet MRI capable of probing in vivo mitochondrial function in intact brain revealed significant reduction of mitochondrial function in the fetal brain after 3Gy TBI. This was validated by ex vivo Oroboros mitochondrial respirometry. Maternal administration JP4-039 one day after TBI (E14.5), which can pass through the placental barrier, significantly reduced fetal brain radiation injury and improved fetal brain mitochondrial respiration. This also preserved cerebral brain tissue integrity and reduced cerebral hemorrhage and cell death. As JP4-039 administration did not change litter sizes or fetus viability, together these findings indicate JP4-039 can be deployed as a safe and effective mitigator of fetal radiation injury from mid-gestational in utero ionizing radiation exposure. One Sentence Summary: Mitochondrial-targeted gramicidin S (GS)-nitroxide JP4-039 is safe and effective radiation mitigator for mid-gestational fetal irradiation injury.

2.
Curr Rev Clin Exp Pharmacol ; 16(3): 263-272, 2021.
Article in English | MEDLINE | ID: mdl-32778037

ABSTRACT

BACKGROUND: To address multidrug resistance, we developed engineered Cationic Antimicrobial Peptides (eCAPs). Lead eCAP WLBU2 displays potent activity against drug-resistant bacteria and effectively treats lethal bacterial infections in mice, reducing bacterial loads to undetectable levels in diverse organs. OBJECTIVE: To support the development of WLBU2, we conducted a mass balance study. METHODS: CD1 mice were administered 10, 15, 20 and 30 mg/kg of QDx5 WLBU2 or a single dose of [14C]-WLBU2 at 15 mg/kg IV. Tolerability, tissue distribution and excretion were evaluated with liquid scintillation and HPLC-radiochromatography. RESULTS: The maximum tolerated dose of WLBU2 is 20 mg/kg IV. We could account for greater than >96% of the radioactivity distributed within mouse tissues at 5 and 15 min. By 24h, only ~40-50% of radioactivity remained in the mice. The greatest % of the dose was present in liver, accounting for ~35% of radioactivity at 5 and 15 min, and ~ 8% of radioactivity remained at 24h. High radioactivity was also present in kidneys, plasma, red blood cells and lungs, while less than 0.2% of radioactivity was present in brain, fat, or skeletal muscle. Urinary and fecal excretion accounted for 12.5 and 2.2% of radioactivity at 24h. CONCLUSION: WLBU2 distributes widely to mouse tissues and is rapidly cleared with a terminal radioactivity half-life of 22 h, a clearance of 27.4 mL/h/kg, and a distribution volume of 0.94 L/kg. At 2-100 µg-eq/g, the concentrations of 14C-WLBU2 appear high enough in the tissues to account for the inhibition of microbial growth.


Subject(s)
Antimicrobial Cationic Peptides , Bacterial Infections , Animals , Antimicrobial Peptides , Carbon Radioisotopes , Mice
3.
SLAS Discov ; 24(6): 653-668, 2019 07.
Article in English | MEDLINE | ID: mdl-31039321

ABSTRACT

Systematic unbiased high-throughput screening (HTS) of drug combinations (DCs) in well-characterized tumor cell lines is a data-driven strategy to identify novel DCs with potential to be developed into effective therapies. Four DCs from a DC HTS campaign were selected for confirmation; only one appears in clinicaltrials.gov and limited preclinical in vitro data indicates that the drug pairs interact synergistically. Nineteen DC-tumor cell line sets were confirmed to interact synergistically in three pharmacological interaction models. We developed an imaging assay to quantify accumulation of the ABCG2 efflux transporter substrate Hoechst. Gefitinib and raloxifene enhanced Hoechst accumulation in ABCG2 (BCRP)-expressing cells, consistent with inhibition of ABCG2 efflux. Both drugs also inhibit ABCB1 efflux. Mitoxantrone, daunorubicin, and vinorelbine are substrates of one or more of the ABCG2, ABCB1, or ABCC1 efflux transporters expressed to varying extents in the selected cell lines. Interactions between ABC drug efflux transporter inhibitors and substrates may have contributed to the observed synergy; however, other mechanisms may be involved. Novel synergistic DCs identified by HTS were confirmed in vitro, and plausible mechanisms of action studied. Similar approaches may justify the testing of novel HTS-derived DCs in mouse xenograft human cancer models and support the clinical evaluation of effective in vivo DCs in patients.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , High-Throughput Screening Assays , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , ATP-Binding Cassette Transporters/genetics , Animals , Cell Culture Techniques , Cell Line, Tumor , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Drug Synergism , Humans , Molecular Imaging , Pilot Projects
4.
SLAS Discov ; 24(3): 242-263, 2019 03.
Article in English | MEDLINE | ID: mdl-30500310

ABSTRACT

Animal and clinical studies demonstrate that cancer drug combinations (DCs) are more effective than single agents. However, it is difficult to predict which DCs will be more efficacious than individual drugs. Systematic DC high-throughput screening (HTS) of 100 approved drugs in the National Cancer Institute's panel of 60 cancer cell lines (NCI-60) produced data to help select DCs for further consideration. We miniaturized growth inhibition assays into 384-well format, increased the fetal bovine serum amount to 10%, lengthened compound exposure to 72 h, and used a homogeneous detection reagent. We determined the growth inhibition 50% values of individual drugs across 60 cell lines, selected drug concentrations for 4 × 4 DC matrices (DCMs), created DCM master and replica daughter plate sets, implemented the HTS, quality control reviewed the data, and analyzed the results. A total of 2620 DCMs were screened in 60 cancer cell lines to generate 3.04 million data points for the NCI ALMANAC (A Large Matrix of Anti-Neoplastic Agent Combinations) database. We confirmed in vitro a synergistic drug interaction flagged in the DC HTS between the vinca-alkaloid microtubule assembly inhibitor vinorelbine (Navelbine) tartrate and the epidermal growth factor-receptor tyrosine kinase inhibitor gefitinib (Iressa) in the SK-MEL-5 melanoma cell line. Seventy-five percent of the DCs examined in the screen are not currently in the clinical trials database. Selected synergistic drug interactions flagged in the DC HTS described herein were subsequently confirmed by the NCI in vitro, evaluated mechanistically, and were shown to have greater than single-agent efficacy in mouse xenograft human cancer models. Enrollment is open for two clinical trials for DCs that were identified in the DC HTS. The NCI ALMANAC database therefore constitutes a valuable resource for selecting promising DCs for confirmation, mechanistic studies, and clinical translation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Screening Assays, Antitumor/methods , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Interactions , High-Throughput Screening Assays , Humans
5.
Sci Transl Med ; 10(427)2018 02 07.
Article in English | MEDLINE | ID: mdl-29437148

ABSTRACT

The gastrointestinal (GI) epithelium is the fastest renewing adult tissue and is maintained by tissue-specific stem cells. Treatment-induced GI side effects are a major dose-limiting factor for chemotherapy and abdominal radiotherapy and can decrease the quality of life in cancer patients and survivors. p53 is a key regulator of the DNA damage response, and its activation results in stimulus- and cell type-specific outcomes via distinct effectors. We demonstrate that p53-dependent PUMA induction mediates chemotherapy-induced intestinal injury in mice. Genetic ablation of Puma, but not of p53, protects against chemotherapy-induced lethal GI injury. Blocking chemotherapy-induced loss of LGR5+ stem cells by Puma KO or a small-molecule PUMA inhibitor (PUMAi) prevents perturbation of the stem cell niche, rapid activation of WNT and NOTCH signaling, and stem cell exhaustion during repeated exposures. PUMAi also protects human and mouse colonic organoids against chemotherapy-induced apoptosis and damage but does not protect cancer cells in vitro or in vivo. Therefore, targeting PUMA is a promising strategy for normal intestinal chemoprotection because it selectively blocks p53-dependent stem cell loss but leaves p53-dependent protective effects intact.


Subject(s)
Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Death/genetics , Cell Death/physiology , Intestines/cytology , Irinotecan/adverse effects , Mice , Mice, Knockout , Signal Transduction/drug effects , Stem Cells/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
J Pharm Biomed Anal ; 150: 169-175, 2018 Feb 20.
Article in English | MEDLINE | ID: mdl-29245086

ABSTRACT

JP4-039 radio-protects prior to, and radio-mitigates after ionizing radiation by neutralizing reactive oxygen species. We developed and validated an LC-MS/MS assay for the quantitation of JP4-039 in murine plasma. Methanol protein precipitation of 50µL plasma was followed by isocratic reverse phase chromatography for a 6min run time, and electrospray positive mode ionization mass spectrometric detection. The plasma assay was linear from 1 to 1000ng/mL with appropriate accuracy (97.1-107.6%) and precision (3.7-12.5%CV), and fulfilled FDA guidance criteria. Recovery was 77.2-136.1% with moderate ionization enhancement (10.9-39.5%). Plasma freeze-thaw stability (98.8-104.2%), stability for 13.5 months at -80°C (93.1-105.6%), and stability for 4h at room temperature (94.2-97.6%) were all acceptable. Limited cross-validation to tissue homogenates suggested that these could also be analyzed for JP4-039 accurately. This assay has been directly applied to determine the pharmacokinetics of JP4-039 in C57BL/6 male mice after IV administration of 20mg/kg JP4-039 and will be extended to other studies of this agent.


Subject(s)
Chromatography, Reverse-Phase , Drug Monitoring/methods , Nitrogen Oxides/blood , Radiation-Protective Agents/metabolism , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Administration, Intravenous , Animals , Calibration , Chromatography, Reverse-Phase/standards , Cold Temperature , Drug Monitoring/standards , Drug Stability , Male , Mice, Inbred C57BL , Nitrogen Oxides/administration & dosage , Nitrogen Oxides/pharmacokinetics , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/pharmacokinetics , Reference Standards , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization/standards , Tandem Mass Spectrometry/standards
7.
J Pharm Biomed Anal ; 146: 154-160, 2017 Nov 30.
Article in English | MEDLINE | ID: mdl-28881312

ABSTRACT

The ribonucleotide reductase inhibitor and radiosensitizer triapine (3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP), NSC 663249) is clinically being evaluated via the intravenous (IV) route for the treatment of cervical and vulvar cancer in combination with primary cisplatin chemoradiation. The need for a 2-h infusion and frequent administration of triapine is logistically challenging, prompting us to pursue oral (PO) administration. In support of the clinical trial investigating oral triapine in combination with chemoradiation, we developed and validated a novel LC-MS/MS assay for the quantification of triapine in 50µL human plasma. After protein precipitation, chromatographic separation of the supernatant was achieved with a Shodex ODP2 column and an isocratic acetonitrile-water mobile phase with 10% ammonium acetate. Detection with an ABI 4000 mass spectrometer utilized electrospray positive mode ionization. The assay was linear from 3 to 3,000ng/mL and proved to be accurate (97.1-103.1%) and precise (<7.4% CV), and met the U.S. FDA guidance for bioanalytical method validation. This LC-MS/MS assay will be an essential tool to further define the pharmacokinetics and oral bioavailability of triapine.


Subject(s)
Enzyme Inhibitors/blood , Enzyme Inhibitors/chemistry , Plasma/chemistry , Pyridines/blood , Pyridines/chemistry , Ribonucleotide Reductases/antagonists & inhibitors , Thiosemicarbazones/blood , Thiosemicarbazones/chemistry , Biological Assay/methods , Chromatography, Liquid/methods , Humans , Reproducibility of Results , Tandem Mass Spectrometry/methods
8.
Lab Anim ; 51(5): 534-537, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28948895

ABSTRACT

Several cancer chemotherapies cause nausea and vomiting, which can be dose-limiting. Musk shrews are used as preclinical models for chemotherapy-induced emesis and for antiemetic effectiveness. Unlike rats and mice, shrews possess a vomiting reflex and demonstrate an emetic profile similar to humans, including acute and delayed phases. As with most animals, dosing of shrews is based on body weight, while translation of such doses to clinically equivalent exposure requires doses based on body surface area. In the current study body surface area in musk shrews was directly assessed to determine the Meeh constant (Km) conversion factor (female = 9.97, male = 9.10), allowing estimation of body surface area based on body weight. These parameters can be used to determine dosing strategies for shrew studies that model human drug exposures, particularly for investigating the emetic liability of cancer chemotherapeutic agents.


Subject(s)
Body Surface Area , Shrews , Vomiting/chemically induced , Animals , Antiemetics , Female , Male , Mice , Rats
9.
Cancer Chemother Pharmacol ; 78(6): 1225-1235, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27778071

ABSTRACT

PURPOSE: The oncogenic transcription factor signal transducer and activator of transcription 3 (STAT3) promotes gene transcription involved in cancer, and its activation by IL-6 is found in head and neck squamous cell carcinoma. Four triazolothiadizine STAT3 pathway inhibitors were evaluated to prioritize a single compound for in vivo examination. METHODS: Metabolic stability in mouse liver microsome incubation was used to evaluate four triazolothiadizine analogues, and UPCDC-10205 was administered to mice IV as single or multiple doses to evaluate toxicity. Single-dose pharmacokinetics (PK), bioavailability and metabolism were studied after IV 4 mg/kg, PO 4 mg/kg, or PO 30 mg/kg suspension in 1% carboxymethyl cellulose. Mice were euthanized between 5 min to 24 h after dosing, and plasma and tissues were analyzed by LC-MS. Non-compartmental PK parameters were determined. RESULTS: Of the four triazolothiadizine analogues evaluated, UPCDC-10205 was metabolically most stable. The maximum soluble dose of 4 mg/kg in 10% Solutol™ was not toxic to mice after single and multiple doses. PK analysis showed extensive tissue distribution and rapid plasma clearance. Bioavailability was ~5%. A direct glucuronide conjugate was identified as the major metabolite which was recapitulated in vitro. CONCLUSIONS: Rapid clearance of UPCDC-10205 was thought to be the result of phase II metabolism despite its favorable stability in a phase I in vitro metabolic stability assay. The direct glucuronidation explains why microsomal stability (reflective of phase I metabolism) did not translate to in vivo metabolic stability. UPCDC-10205 did not demonstrate appropriate exposure to support efficacy studies in the current formulation.


Subject(s)
Interleukin-6/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Thiadiazines/pharmacokinetics , Triazoles/pharmacokinetics , Animals , Female , Mice , Microsomes, Liver/metabolism , Thiadiazines/toxicity , Triazoles/toxicity
10.
J Pharm Biomed Anal ; 129: 359-366, 2016 Sep 10.
Article in English | MEDLINE | ID: mdl-27454087

ABSTRACT

The hypomethylating agent 5-fluoro-2'-deoxycytidine (FdCyd, NSC 48006) is being evaluated clinically both via the intravenous route and via the oral route in combination with 3,4,5,6-tetrahydrouridine (THU), a potent inhibitor of FdCyd catabolism. To determine the pharmacokinetics of FdCyd and downstream metabolites, we developed and validated an LC-MS/MS assay for the quantitation of FdCyd, 5-fluoro-2'-deoxyuridine (FdUrd), and 5-fluorouracil (FU) in 0.2mL human plasma. After acetonitrile protein precipitation, the sample was split and separate chromatography was achieved for FdCyd with a Synergi Polar-RP column and for FdUrd and FU with a Shodex Asahipak NH2P-50 2D column. Gradients of 0.1% acetic acid in acetonitrile and water were used. Detection with a Quattromicro quadrupole mass spectrometer with electrospray ionization in positive-ion (FdCyd) or negative-ion (FdUrd and FU) multiple reaction monitoring (MRM) mode. The assay was linear from 5 to 3000ng/mL for all three analytes and proved to be accurate (96.7-105.5%) and precise (<8.1%CV), and fulfilled FDA criteria for bioanalytical method validation. We demonstrated the suitability of this assay for measuring FdCyd and metabolites FdUrd and FU in plasma from a patient who was administered 120mg PO FdCyd 30min after 3000mg THU. Our LC-MS/MS assay will be an essential tool to further define the pharmacology of FdCyd in ongoing and future studies.


Subject(s)
Deoxyuridine/analogs & derivatives , Fluorouracil/blood , Fluorouracil/chemistry , Plasma/chemistry , Biological Assay/methods , Chromatography, Liquid/methods , Deoxyuridine/blood , Deoxyuridine/chemistry , Deoxyuridine/pharmacokinetics , Humans , Tandem Mass Spectrometry/methods , Tetrahydrouridine/chemistry
11.
Cancer Chemother Pharmacol ; 78(1): 73-81, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27169793

ABSTRACT

PURPOSE: Indenoisoquinolines are non-camptothecin topoisomerase I (TopI) inhibitors that overcome the limitations of camptothecins: chemical instability and camptothecin resistance. Two dosing schedules of the novel indenoisoquinoline, indotecan (LMP400), were evaluated in patients with advanced solid tumors. METHODS: The maximum tolerated dose (MTD), toxicities, and pharmacokinetics of two indotecan drug administration schedules (daily for 5 days or weekly) were investigated. Modulation of TopI and the phosphorylation of histone H2AX (γH2AX) were assayed in tumor biopsies; γH2AX levels were also evaluated in circulating tumor cells (CTCs) and hair follicles to assess DNA damage response. RESULTS: An MTD of 60 mg/m(2)/day was established for the daily regimen, compared to 90 mg/m(2) for the weekly regimen. The TopI response to drug showed target engagement in a subset of tumor biopsies. Pharmacokinetics profiles demonstrated a prolonged terminal half-life and tissue accumulation compared to topotecan. Dose-dependent decreases in total CTCs were measured in seven patients. Formation of γH2AX-positive foci in CTCs (day 3) and hair follicles (4-6 h) was observed following treatment. CONCLUSIONS: We established the MTD of two dosing schedules for a novel TopI inhibitor, indotecan. Target engagement was demonstrated as Top1 downregulation and γH2AX response. No objective responses were observed on either schedule in this small patient cohort. The principal toxicity of both schedules was myelosuppression; no significant gastrointestinal problems were observed. Increased DNA damage response was observed in CTCs, hair follicles, and a subset of tumor biopsies.


Subject(s)
Benzodioxoles/administration & dosage , DNA Topoisomerases, Type I/metabolism , Histones/metabolism , Isoquinolines/administration & dosage , Neoplasms/drug therapy , Topoisomerase I Inhibitors/administration & dosage , Adult , Aged , Benzodioxoles/adverse effects , Benzodioxoles/pharmacokinetics , DNA Damage/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Administration Schedule , Female , Hair Follicle/metabolism , Half-Life , Humans , Isoquinolines/adverse effects , Isoquinolines/pharmacokinetics , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/pathology , Time Factors , Tissue Distribution , Topoisomerase I Inhibitors/adverse effects , Topoisomerase I Inhibitors/pharmacokinetics , Topotecan/pharmacokinetics , Young Adult
12.
Adv Biol Chem ; 6(6): 193-215, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28890844

ABSTRACT

BACKGROUND: Cervical cancer is the second most common cancer in women worldwide [1]. Photodynamic therapy has been used for cervical intraepithelial neoplasia with good responses, but few studies have used newer phototherapeutics. We evaluated the effectiveness of photodynamic therapy using Pc 4 in vitro and in vivo against human cervical cancer cells. METHODS: CaSki and ME-180 cancer cells were grown as monolayers and spheroids. Cell growth and cytotoxicity were measured using a methylthiazol tetrazolium assay. Pc 4 cellular uptake and intracellular distrubtion were determined. For in vitro Pc 4 photodynamic therapy cells were irradiated at 667nm at a fluence of 2.5 J/cm2 at 48 h. SCID mice were implanted with CaSki and ME-180 cells both subcutaneously and intracervically. Forty-eight h after Pc 4 photodynamic therapy was administered at 75 and 150 J/cm2. RESULTS: The IC50s for Pc 4 and Pc 4 photodynamic therapy for CaSki and ME-180 cells as monolayers were, 7.6µM and 0.016µM and >10µM and 0.026µM; as spheroids, IC50s of Pc 4 photodynamic therapy were, 0.26µM and 0.01µM. Pc 4 was taken up within cells and widely distributed in tumors and tissues. Intracervical photodynamic therapy resulted in tumor death, however mice died due to gastrointestinal toxicity. Photodynamic therapy resulted in subcutaneous tumor death and growth delay. CONCLUSIONS: Pc 4 photodynamic therapy caused death within cervical cancer cells and xenografts, supporting development of Pc 4 photodynamic therapy for treatment of cervical cancer. Support: P30-CA47904, CTSI BaCCoR Pilot Program.

13.
Cancer Chemother Pharmacol ; 76(4): 803-11, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26321472

ABSTRACT

INTRODUCTION: 5-Fluoro-2'-deoxycytidine (FdCyd; NSC48006), a fluoropyrimidine nucleoside inhibitor of DNA methylation, is degraded by cytidine deaminase (CD). Pharmacokinetic evaluation was carried out in cynomolgus monkeys in support of an ongoing phase I study of the PO combination of FdCyd and the CD inhibitor tetrahydrouridine (THU; NSC112907). METHODS: Animals were dosed intravenously (IV) or per os (PO). Plasma samples were analyzed by LC-MS/MS for FdCyd, metabolites, and THU. Clinical chemistry and hematology were performed at various times after dosing. A pilot pharmacokinetic study was performed in humans to assess FdCyd bioavailability. RESULTS: After IV FdCyd and THU administration, FdCyd C(max) and AUC increased with dose. FdCyd half-life ranged between 22 and 56 min, and clearance was approximately 15 mL/min/kg. FdCyd PO bioavailability after THU ranged between 9 and 25 % and increased with increasing THU dose. PO bioavailability of THU was less than 5 %, but did result in plasma concentrations associated with inhibition of its target CD. Human pilot studies showed comparable bioavailability for FdCyd (10 %) and THU (4.1 %). CONCLUSION: Administration of THU with FdCyd increased the exposure to FdCyd and improved PO FdCyd bioavailability from <1 to 24 %. Concentrations of THU and FdCyd achieved after PO administration are associated with CD inhibition and hypomethylation, respectively. The schedule currently studied in phase I studies of PO FdCyd and THU is daily times three at the beginning of the first and second weeks of a 28-day cycle.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Cytidine Deaminase/antagonists & inhibitors , Deoxycytidine/analogs & derivatives , Enzyme Inhibitors/pharmacokinetics , Tetrahydrouridine/pharmacokinetics , Administration, Oral , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/blood , Biological Availability , Biotransformation , Cohort Studies , Deoxycytidine/administration & dosage , Deoxycytidine/blood , Deoxycytidine/pharmacokinetics , Dose-Response Relationship, Drug , Drug Combinations , Drug Evaluation, Preclinical , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/blood , Female , Half-Life , Humans , Infusions, Intravenous , Macaca fascicularis , Male , Metabolic Clearance Rate , Pilot Projects , Tetrahydrouridine/administration & dosage , Tetrahydrouridine/blood
14.
Cancer Chemother Pharmacol ; 76(3): 537-46, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26193988

ABSTRACT

INTRODUCTION: Benzaldehyde dimethane sulfonate (BEN, DMS612, NSC281612) is a bifunctional alkylating agent currently in clinical trials. We previously characterized the degradation products of BEN in plasma and blood. The conversion of BEN to its carboxylic acid analogue (BA) in whole blood, but not plasma, suggests that an enzyme in RBCs may be responsible for this conversion. BEN conversion to BA was observed in renal carcinoma cells and appeared to correlate with IC50. To better understand the pharmacology of BEN, we aimed to evaluate the metabolism and enzymes potentially responsible for the conversion of BEN to BA. METHODS: Human red blood cells (RBC) were used to characterize kinetics and susceptibility to enzyme-specific inhibitors. Recombinant enzymes were used to confirm metabolism of BEN to BA. Analytes were quantitated with established LC-MS/MS methods. RESULTS: Average apparent Vmax and Km were 68 ng/mL min(-1) [10% RBC](-1) and 373 ng/mL, respectively. The conversion of BEN to BA in RBC was not inhibited by carbon monoxide, nitrogen gas, or menadione, an inhibitor of aldehyde oxidase. The conversion was inhibited by disulfiram, an inhibitor of ALDH. Of available ALDH isoforms ALDH1A1, ALDH3A1, ALDH2, and ALDH5A1, only ALDH1A1 converted BEN to BA. CONCLUSION: The activating conversion of BEN to BA is mediated not by CYP450 enzymes or aldehyde oxidase, but by ALDH1A1. This enzyme, a potential stem cell marker, may be a candidate biomarker for clinical activity of BEN.


Subject(s)
Antineoplastic Agents, Alkylating/blood , Benzaldehydes/blood , Carcinoma, Renal Cell/blood , Kidney Neoplasms/blood , Carcinoma, Renal Cell/enzymology , Cell Line, Tumor , Chromatography, High Pressure Liquid , Erythrocytes/enzymology , Erythrocytes/metabolism , Humans , Immunoprecipitation
15.
Cancer Chemother Pharmacol ; 76(2): 287-99, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26050209

ABSTRACT

PURPOSE: The interaction of p53 with its negative regulators Mdm2/4 has been widely studied (Khoury and Domling in Curr Pharm Des 18(30):4668-4678, 2012). In p53(+/+) cells, expression of Mdm2/4 leads to p53 turnover, inhibition of downstream transcription, decreasing cell cycle arrest, or apoptosis. We report in vitro cytotoxicity and in vivo efficacy, pharmacokinetics, and metabolism of YH264, YH263, and WW751, three proposed small molecule inhibitors of the Mdm2/4-p53 interaction. METHODS: MTT cytotoxicity assays were performed, and alterations in proteins were examined using western blots. Mice were dosed 150 mg/kg YH264 or YH263 IV or PO QDx5. Mice were IV dosed 88, 57, or 39 mg/kg WW751 for 3, 5, or 5 days. YH264, YH263, and WW751 and metabolites were quantitated by LC-MS/MS. RESULTS: IC50 values for YH264, YH263, and WW751 against p53 wild-type HCT 116 cells after 72 h of incubation were 18.3 ± 2.3, 8.9 ± 0.6, and 3.1 ± 0.2 µM, respectively. Only YH264 appeared to affect p53 expression in vitro. None of the compounds affected the growth of HCT 116 xenografts in C.B-17 SCID mice. YH264 plasma half-life was 147 min; YH263 plasma half-life was 263 min; and WW751 plasma half-life was less than 120 min. CONCLUSIONS: Despite dosing the mice at the maximum soluble doses, we could not achieve tumor concentrations equivalent to the intracellular concentrations required to inhibit cell growth in vitro. YH263 and WW751 do not appear to affect p53/Mdm2, and none of the three were active in a subcutaneous HCT 116 p53(+/+) xenograft model.


Subject(s)
Antineoplastic Agents/chemistry , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins/metabolism , Pyrazoles/chemistry , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Female , HCT116 Cells , Heterografts , Humans , Mice, SCID , Neoplasm Transplantation , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology
16.
Cancer Chemother Pharmacol ; 75(5): 1015-23, 2015 May.
Article in English | MEDLINE | ID: mdl-25776905

ABSTRACT

PURPOSE: NSC 743400 is a novel synthetic indenoisoquinoline analog under development as an anticancer agent. It is a potent topoisomerase I inhibitor with potential therapeutic advantages over FDA-approved camptothecin derivatives. In preparation for clinical development of NSC 743400, we determined the pharmacokinetics after administration to rats and dogs. METHODS: NSC 743400 was administered intravenously at a dose of 12 or 24 mg/m(2) to rats (single bolus) or 10, 50, 100, 215, 430, or 646 mg/m(2) (intravenous infusion) or 860 or 1720 mg/m(2) (orally) to dogs. RESULTS: Intravenously administered NSC 743400 was eliminated from both species with an estimated t 1/2 of 2-5 h in rat and 6-14 h in dog. Elimination t 1/2 increased with dose in dog. Area under the plasma concentration-versus-time curve (AUC) was comparable in both species, at about 300-400 h ng/mL for the approximately 10 mg/m(2) dose groups. Overall, AUC values increased proportionally with dose for both species but had evidence of more than proportional exposure at the highest doses. Oral dosing resulted in variable drug absorption. CONCLUSIONS: The pharmacokinetic data were used to plan first-in-human clinical trials.


Subject(s)
Benzodioxoles/blood , Isoquinolines/blood , Topoisomerase I Inhibitors/blood , Animals , Benzodioxoles/administration & dosage , Benzodioxoles/pharmacokinetics , Dogs , Dose-Response Relationship, Drug , Female , Infusions, Intravenous , Injections, Intravenous , Isoquinolines/administration & dosage , Isoquinolines/pharmacokinetics , Male , Random Allocation , Rats , Rats, Inbred F344 , Topoisomerase I Inhibitors/administration & dosage , Topoisomerase I Inhibitors/pharmacokinetics
17.
Cancer Chemother Pharmacol ; 75(3): 537-46, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25567350

ABSTRACT

PURPOSE: Inhibitors of DNA (cytosine-5)-methyltransferases (DNMT) are active antineoplastic agents. We conducted the first-in-human phase I trial of 5-fluoro-2'-deoxycytidine (FdCyd), a DNMT inhibitor stable in aqueous solution, in patients with advanced solid tumors. Objectives were to establish the safety, maximum tolerated dose (MTD), pharmacokinetics, and pharmacodynamics of FdCyd + tetrahydrouridine (THU). METHODS: FdCyd + THU were administered by 3 h IV infusion on days 1-5 every 3 weeks, or days 1-5 and 8-12 every 4 weeks. FdCyd was administered IV with a fixed 350 mg/m(2)/day dose of THU to inhibit deamination of FdCyd. Pharmacokinetics of FdCyd, downstream metabolites and THU were assessed by LC-MS/MS. RBC γ-globin expression was evaluated as a pharmacodynamics biomarker. RESULTS: Patients were enrolled on the 3-week schedule at doses up to 80 mg/m(2)/day without dose-limiting toxicity (DLT) prior to transitioning to the 4-week schedule, which resulted in an MTD of 134 mg/m(2)/day; one of six patients had a first-cycle DLT (grade 3 colitis). FdCyd ≥40 mg/m(2)/day produced peak plasma concentrations >1 µM. Although there was inter-patient variability, γ-globin mRNA increased during the first two treatment cycles. One refractory breast cancer patient experienced a partial response (PR) of >90 % decrease in tumor size, lasting over a year. CONCLUSIONS: The MTD was established at 134 mg/m(2) FdCyd + 350 mg/m(2) THU days 1-5 and 8-12 every 4 weeks. Based on toxicities observed over multiple cycles, good plasma exposures, and the sustained PR observed at 67 mg/m(2)/day, the phase II dose for our ongoing multi-histology trial is 100 mg/m(2)/day FdCyd with 350 mg/m(2)/day THU.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Neoplasms/drug therapy , gamma-Globulins/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chromatography, Liquid , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/pathology , Tandem Mass Spectrometry , Tetrahydrouridine/administration & dosage , Treatment Outcome
18.
Cancer Chemother Pharmacol ; 75(1): 143-52, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25398697

ABSTRACT

PURPOSE: Cisplatin induces nausea and emesis, even with antiemetic supportive care. To assess platinum exposure, which could activate nausea and emesis, we quantitated platinum in the brain and various organs, and hindbrain and spinal cord substance P, a key neuropeptide for the neuronal signaling of nausea and emesis. METHODS: Musk shrews, a model species for nausea and emesis research, were dosed intraperitoneally with 20 mg/kg cisplatin and euthanized at up to 72 h after injection. Concentrations of platinum were quantitated in plasma ultrafiltrate, plasma, lung, kidney, combined forebrain and midbrain, hindbrain, and spinal cord by flameless atomic absorption spectrometry. Hindbrains and spinal cords were analyzed for substance P by immunohistochemistry after injection of 20 or 30 mg/kg. RESULTS: Plasma ultrafilterable platinum concentrations decreased rapidly till 60 min after dosing and then more slowly by 24 h. The concentrations of total platinum in both the fore- and midbrain and the hindbrain were similar at all time points and were at least 20-fold lower than plasma total platinum concentrations. There were no significant changes in substance P immunoreactivity after cisplatin dosing. Histology revealed damage to the renal cortex by 72 h after injection of cisplatin. CONCLUSIONS: This is the first study to examine platinum concentrations in musk shrews after administration of cisplatin and delineate substance P immunohistochemical staining in the hindbrain and spinal cord of this species. The platinum concentrations detected in the brain could potentially contribute to the neurological side effects of cisplatin, such as nausea and emesis.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cisplatin/pharmacokinetics , Disease Models, Animal , Emetics/pharmacokinetics , Nausea/chemically induced , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/drug effects , Brain/metabolism , Brain/pathology , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cisplatin/administration & dosage , Cisplatin/adverse effects , Cisplatin/blood , Dose-Response Relationship, Drug , Emetics/administration & dosage , Emetics/adverse effects , Emetics/blood , Female , Half-Life , Injections, Intraperitoneal , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Lung/drug effects , Lung/metabolism , Lung/pathology , Nausea/blood , Nausea/metabolism , Nausea/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Platinum/blood , Platinum/metabolism , Shrews , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , Substance P/metabolism , Tissue Distribution
19.
Cancer Chemother Pharmacol ; 72(6): 1195-204, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24061865

ABSTRACT

PURPOSE: Benzaldehyde dimethane sulfonate (DMS612, NSC281612, BEN) is an alkylator with activity against renal cell carcinoma, currently in phase I trials. In blood, BEN is rapidly metabolized into its highly reactive carboxylic acid (BA), presumably the predominant alkylating species. We hypothesized that BEN is metabolized to BA by aldehyde dehydrogenase (ALDH) and aimed to increase BEN exposure in blood and tissues by inhibiting ALDH with disulfiram, thereby shifting BA production from blood to tissues. METHODS: Female CD2F1 mice were dosed with 20 mg/kg BEN iv alone or 24 h after 300 mg/kg disulfiram ip. BEN, BA, and metabolites were quantitated in plasma and urine, and toxicities were assessed. RESULTS: BEN had a plasma t½ <5 min and produced at least 12 products. The metabolite half-lives were <136 min. Disulfiram increased BEN plasma exposure 368-fold (AUC0-inf from 0.11 to 40.5 mg/L min), while plasma levels of BA remained similar. Urinary BEN excretion increased (1.0-1.5 % of dose), while BA excretion was unchanged. Hematocrit, white blood cell counts, and percentage lymphocytes decreased after BEN administration. Coadministration of disulfiram appeared to enhance these effects. Profound liver pathology was observed in mice treated with disulfiram and BEN. CONCLUSIONS: BEN plasma concentrations increased after administration of disulfiram, suggesting that ALDH mediates the rapid metabolism of BEN in vivo, which may explain the increased toxicity seen with BEN after administration of disulfiram. Our results suggest that the coadministration of BEN with drugs that inhibit ALDH to patients that are ALDH deficient may cause liver damage.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Antineoplastic Agents/pharmacokinetics , Benzaldehydes/pharmacokinetics , Disulfiram/pharmacology , Enzyme Inhibitors/pharmacology , Animals , Antineoplastic Agents/toxicity , Area Under Curve , Benzaldehydes/toxicity , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Drug Interactions , Female , Half-Life , Mice
20.
Mol Pharm ; 10(5): 1988-97, 2013 May 06.
Article in English | MEDLINE | ID: mdl-23531079

ABSTRACT

Photodynamic therapy (PDT) holds great promise for the treatment of head and neck (H&N) carcinomas where repeated loco-regional therapy often becomes necessary due to the highly aggressive and recurrent nature of the cancers. While interstitial light delivery technologies are being refined for PDT of H&N and other cancers, a parallel clinically relevant research area is the formulation of photosensitizers in nanovehicles that allow systemic administration yet preferential enhanced uptake in the tumor. This approach can render dual-selectivity of PDT, by harnessing both the drug and the light delivery within the tumor. To this end, we report on a cell-targeted nanomedicine approach for the photosensitizer silicon phthalocyanine-4 (Pc 4), by packaging it within polymeric micelles that are surface-decorated with GE11-peptides to promote enhanced cell-selective binding and receptor-mediated internalization in EGFR-overexpressing H&N cancer cells. Using fluorescence spectroscopy and confocal microscopy, we demonstrate in vitro that the EGFR-targeted Pc 4-nanoformulation undergoes faster and higher uptake in EGFR-overexpressing H&N SCC-15 cells. We further demonstrate that this enhanced Pc 4 uptake results in significant cell-killing and drastically reduced post-PDT clonogenicity. Building on this in vitro data, we demonstrate that the EGFR-targeted Pc 4-nanoformulation results in significant intratumoral drug uptake and subsequent enhanced PDT response, in vivo, in SCC-15 xenografts in mice. Altogether our results show significant promise toward a cell-targeted photodynamic nanomedicine for effective treatment of H&N carcinomas.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Photochemotherapy/methods , Animals , Biological Transport, Active , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Chemistry, Pharmaceutical , Drug Delivery Systems , ErbB Receptors/metabolism , Female , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Indoles/administration & dosage , Indoles/pharmacokinetics , Mice , Mice, SCID , Nanomedicine/methods , Nanoparticles/administration & dosage , Organosilicon Compounds/administration & dosage , Organosilicon Compounds/pharmacokinetics , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacokinetics , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...