Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Chromatogr Sci ; 57(3): 195-203, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30395213

ABSTRACT

The objective of this study was to validate a method for the determination of laromustine (VNP40101M) and short-lived its active metabolite (VNP4090CE) that has a half-life in human blood of <90 s in human plasma by liquid chromatography (LC) with tandem mass spectrometric (MS/MS) detection. We overcome the stability dilemma by acidified the human plasma with citric acid. Laromustine "breaks" down on the source of mass spectrometry to give m/z 249 which is the same m/z for VNP4090CE. Because VNP4090CE and laromustine elute at approximate retention time of 1.93 and 2.94 min, respectively, we were able to quantify both of them in one method. VNP40101M, VNP4090CE and the internal standards were extracted from human plasma by liquid-liquid extraction into ethyl ether. The ethyl ether layer was evaporated, reconstituted and analyzed using LC with MS/MS detection. Validation parameters such as selectivity, limit of quantitation, linearity, precision, accuracy, recovery, autosampler viability, freeze-thaw cycles and compounds stability are evaluated for this method. Results were calculated using peak area ratios, and calibration curves were generated using a weighted (1/x2) linear least-squares regression. Calibration curves for VNP40101M and VNP4090CE in human plasma ranged from 1.00 to 1,000 ng/mL. In this study, both intra- and inter-assay results demonstrated a relative standard deviation for calibration standards (inter-assay) and quality control samples (intra- and inter-assay) to be ≤15.0%. In this method, there is ~1.79% isotopic interference of VNP40101M to VNP40101M-IS, and ~3.76% isotopic interference of VNP4090CE to VNP4090CE-IS. It was concluded that there was no significant carryover.


Subject(s)
Chromatography, Liquid/methods , Hydrazines/blood , Sulfonamides/blood , Tandem Mass Spectrometry/methods , Humans , Hydrazines/chemistry , Limit of Detection , Linear Models , Reproducibility of Results , Sulfonamides/chemistry
2.
Xenobiotica ; 47(5): 394-407, 2017 May.
Article in English | MEDLINE | ID: mdl-27440490

ABSTRACT

1. Alkylating agents are capable of introducing an alkyl group into nucleophilic sites on DNA or RNA through covalent bond. Laromustine is an active member of a relatively new class of sulfonylhydrazine prodrugs under development as antineoplastic alkylating agents, and displays significant single-agent activity. 2. This is the first report of the population pharmacokinetic analysis of laromustine, 106 patients, 66 with hematologic malignancies and 40 with solid tumors, participated in five clinical trials worldwide. Of these, 104 patients were included in the final NONMEM analysis. 3. The population estimates for total clearance (CL) and volume of distribution of the central compartment (V1) were 96.3 L/h and 45.9 L, associated with high inter-patient variability of 52.9% and 79.8% and inter-occasion variability of 26.7% and 49.3%, respectively. The population estimates for Q and V2 were 73.2 L/h and 29.9 L, and inter-patient variability in V2 was 63.1%, respectively. 4. The estimate of Vss (75.8 L) exceeds total body water, indicating that laromustine is distributed to tissues. The half-life is short, less than 1 h, reflecting rapid clearance. Population PK analysis showed laromustine pharmacokinetics to be independent of dose and organ function with no effect on subsequent dosing cycles.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Hydrazines/pharmacokinetics , Sulfonamides/pharmacokinetics , Female , Humans , Male , Middle Aged , Models, Biological , Prodrugs
3.
Drug Metab Dispos ; 44(8): 1349-63, 2016 08.
Article in English | MEDLINE | ID: mdl-27278961

ABSTRACT

This review highlights the recent research into the biotransformations and rearrangement of the sulfonylhydrazine-alkylating agent laromustine. Incubation of [(14)C]laromustine with rat, dog, monkey, and human liver microsomes produced eight radioactive components (C-1 to C-8). There was little difference in the metabolite profile among the species examined, partly because NADPH was not required for the formation of most components, which instead involved decomposition and/or hydrolysis. The exception was C-7, a hydroxylated metabolite, largely formed by CYP2B6 and CYP3A4/5. Liquid chromatography-multistage mass spectrometry (LC-MS(n)) studies determined that collision-induced dissociation, and not biotransformation or enzyme catalysis, produced the unique mass spectral rearrangement. Accurate mass measurements performed with a Fourier-transform ion cyclotron resonance mass spectrometer (FTICR-MS) significantly aided determination of the elemental compositions of the fragments and in the case of laromustine revealed the possibility of rearrangement. Further, collision-induced dissociation produced the loss of nitrogen (N2) and methylsulfonyl and methyl isocyanate moieties. The rearrangement, metabolite/decomposition products, and conjugation reactions were analyzed utilizing hydrogen-deuterium exchange, exact mass, (13)C-labeled laromustine, nuclear magnetic resonance spectroscopy (NMR), and LC-MS(n) experiments to assist with the assignments of these fragments and possible mechanistic rearrangement. Such techniques produced valuable insights into these functions: 1) Cytochrome P450 is involved in C-7 formation but plays little or no role in the conversion of [(14)C]laromustine to C-1 through C-6 and C-8; 2) the relative abundance of individual degradation/metabolite products was not species-dependent; and 3) laromustine produces several reactive intermediates that may produce the toxicities seen in the clinical trials.


Subject(s)
Antineoplastic Agents, Alkylating/metabolism , Hydrazines/metabolism , Microsomes, Liver/enzymology , Sulfonamides/metabolism , Animals , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/toxicity , Biotransformation , Cytochrome P-450 CYP2B6/metabolism , Cytochrome P-450 CYP3A/metabolism , Dogs , Haplorhini , Humans , Hydrazines/chemistry , Hydrazines/toxicity , Hydroxylation , Molecular Structure , Rats , Substrate Specificity , Sulfonamides/chemistry , Sulfonamides/toxicity
4.
Xenobiotica ; 45(8): 711-21, 2015.
Article in English | MEDLINE | ID: mdl-25798740

ABSTRACT

1. Laromustine (VNP40101M, also known as Cloretazine) is a novel sulfonylhydrazine alkylating (anticancer) agent. This article describes the use of quantitative whole-body autoradiography (QWBA) and mass balance to study the tissue distribution, the excretion mass balance and pharmacokinetics after intravenous administration of [(14)C]VNP40101M to rats. A single 10 mg/kg IV bolus dose of [(14)C]VNP40101M was given to rats. 2. The recovery of radioactivity from the Group 1 animals over a 7-day period was an average of 92.1% of the administered dose, which was accounted for in the excreta and carcass. Most of the radioactivity was eliminated within 48 h via urine (48%), with less excreted in feces (5%) and expired air accounted for (11%). The plasma half-life of [(14)C]laromustine was approximately 62 min and the peak plasma concentration (Cmax) averaged 8.3 µg/mL. 3. The QWBA study indicated that the drug-derived radioactivity was widely distributed to tissues through 7 days post-dose after a single 10 mg/kg IV bolus dose of [(14)C]VNP40101M to male pigmented Long-Evans rats. The maximum concentrations were observed at 0.5 or 1 h post-dose for majority tissues (28 of 42). The highest concentrations of radioactivity were found in the small intestine contents at 0.5 h (112.137 µg equiv/g), urinary bladder contents at 3 h (89.636 µg equiv/g) and probably reflect excretion of drug and metabolites. The highest concentrations in specific organs were found in the renal cortex at 1 h (28.582 µg equiv/g), small intestine at 3 h (16.946 µg equiv/g), Harderian gland at 3 h (12.332 µg equiv/g) and pancreas at 3 h (12.635 µg equiv/g). Concentrations in the cerebrum (1.978 µg equiv/g), cerebellum (2.109 µg equiv/g), medulla (1.797 µg equiv/g) and spinal cord (1.510 µg equiv/g) were maximal at 0.5 h post-dose and persisted for 7 days. 4. The predicted total body and target organ exposures for humans given a single 100 µCi IV dose of [(14)C]VNP40101M were well within the medical guidelines for maximum radioactivity exposures in human subjects.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Hydrazines/administration & dosage , Hydrazines/pharmacokinetics , Metalloporphyrins/chemistry , Neoplasms/drug therapy , Sulfonamides/administration & dosage , Sulfonamides/pharmacokinetics , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/urine , Humans , Hydrazines/blood , Hydrazines/urine , Injections, Intravenous , Male , Models, Animal , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Sulfonamides/blood , Sulfonamides/urine , Tissue Distribution
5.
Methods Mol Biol ; 542: 649-59, 2009.
Article in English | MEDLINE | ID: mdl-19565926

ABSTRACT

The ideal anticancer regimen is one that is specific for cancer cells with limited toxicity to normal tissues. Genetically modified, nonpathogenic Salmonella offer a potential way to induce direct tumoricidal activity or to deliver tumoricidal agents to tumors. An attenuated strain of Salmonella typhimurium, called VNP20009, and its derivative TAPET-CD (which expresses Escherichia coli cytosine deaminase) are highly selective for tumor tissue and can deliver therapeutic proteins preferentially to tumors in preclinical models. Both VNP20009 and TAPET-CD have been investigated successfully in Phase 1 clinical trials in cancer patients.


Subject(s)
Cytosine Deaminase/metabolism , Genetic Therapy/methods , Neoplasms/therapy , Salmonella typhimurium/genetics , Animals , Chromosomes, Bacterial/genetics , Cloning, Molecular , Escherichia coli/enzymology , Female , Fluorouracil/pharmacokinetics , Genetic Vectors/genetics , Mice , Mice, Inbred C57BL , Organ Specificity , Tissue Distribution
6.
Drug Metab Dispos ; 37(9): 1922-30, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19520774

ABSTRACT

Laromustine (VNP40101M, also known as Cloretazine) is a novel sulfonylhydrazine alkylating (anticancer) agent. Laromustine generates two types of reactive intermediates: 90CE and methylisocyanate. When incubated with rat, dog, monkey, and human liver microsomes, [(14)C]laromustine was converted to 90CE (C-8) and seven other radioactive components (C-1-C-7). There was little difference in the metabolite profile among the species examined, in part because the formation of most components (C-1-C-6 and 90CE) did not require NADPH but involved decomposition and/or hydrolysis. The exception was C-7, a hydroxylated metabolite, largely formed by CYP2B6 and CYP3A4/5. Laromustine caused direct inhibition of CYP2B6 and CYP3A4/5 (the two enzymes involved in C-7 formation) as well as of CYP2C19. K(i) values were 125 microM for CYP2B6, 297 muM for CYP3A4/5, and 349 microM for CYP2C19 and were greater than the average clinical plasma C(max) of laromustine (25 microM). There was evidence of time-dependent inhibition of CYP1A2, CYP2B6, and CYP3A4/5. Treatment of primary cultures of human hepatocytes with up to 100 microM laromustine did not induce CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, or CYP3A4/5, but the highest concentration of laromustine decreased the activity and levels of immunoreactive CYP3A4. The results of this study suggest the laromustine has 1) negligible victim potential with respect to metabolism by cytochrome P450 enzymes, 2) negligible enzyme-inducing potential, and 3) the potential in some cases to cause inhibition of CYP2B6, CYP3A4, and possibly CYP2C19 during and shortly after the duration of intravenous administration of this anticancer drug, but the clinical effects of such interactions are likely to be insignificant.


Subject(s)
Antineoplastic Agents/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/biosynthesis , Hydrazines/pharmacology , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Dogs , Drug Interactions , Enzyme Induction/drug effects , Haplorhini , Humans , Hydrazines/pharmacokinetics , Hydroxylation , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Isoenzymes/biosynthesis , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , NADP/metabolism , Phenotype , Rats , Sulfonamides/pharmacokinetics
7.
Clin Cancer Res ; 14(13): 4259-66, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18594008

ABSTRACT

PURPOSE: We engineered the oncolytic Salmonella typhimurium-derived bacterium VNP20009 as a vector to target delivery to tumors of the prodrug-activating enzyme carboxypeptidase G2 (CPG2) and to show enhanced antitumor efficacy on administration of different prodrugs. EXPERIMENTAL DESIGN: We characterized CPG2 expression in vectors by immunoblotting, immunofluorescence, and enzyme activity. We assessed prodrug activation by high-performance liquid chromatography. Target human tumor cell and bacterial vector cell cytotoxicity was measured by flow cytometry and colony-forming assays. Therapy was shown in two human tumor xenografts and one mouse allograft with postmortem analysis of bacterial and CPG2 concentration in the tumors. RESULTS: CPG2 is expressed within the bacterial periplasm. It activates prodrugs and induces cytotoxicity in human tumor cells but not in host bacteria. Following systemic administration, bacteria multiply within xenografts reaching 2 x 10(7)/g to 2 x 10(8)/g at 40 days postinoculation. The concentration of CPG2 in these tumors increases steadily to therapeutic levels of 1 to 6 units/g. The bacteria alone reduce the growth of the tumors. Subsequent administration of prodrugs further reduces significantly the growth of the xenografts. CONCLUSIONS: The bacteria multiply within tumors, resulting in a selective expression of CPG2. The CPG2-expressing bacteria alone reduce the growth of tumors. However, in the presence of prodrugs activated by CPG2, this oncolytic effect is greatly increased. We conclude that bacterial oncolytic therapy, combined with CPG2-mediated prodrug activation, has great potential in the treatment of a range of cancers.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/therapy , Genetic Therapy/methods , Melanoma/drug therapy , Melanoma/genetics , Prodrugs/metabolism , Salmonella typhimurium/metabolism , gamma-Glutamyl Hydrolase/genetics , Animals , Cell Line, Tumor , Chromatography, High Pressure Liquid , Flow Cytometry , Genetic Vectors , Humans , Mice , Neoplasm Transplantation
8.
Neuro Oncol ; 9(3): 240-4, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17522334

ABSTRACT

VNP40101M, or 1,2-bis(methylsulfonyl)-1-(2-choloroethyl)-2-(methylamino)carbonylhydrazine (Cloretazine), is a bifunctional prodrug that belongs to a class of DNA-modifying agents-the sulfonylhydrazines-that has been synthesized and been shown to have activity against a wide spectrum of xenografts. The current study was designed to assess the activity of VNP40101M administered at a dose of 18 mg/kg daily for five days against a panel of human adult and pediatric CNS tumors growing subcutaneously or intracranially in athymic nude mice. The results demonstrated statistically significant (p < 0.05) growth delays of 15.0, 8.3, 51.0, 60+, 60+, and 60+ days in subcutaneous xenografts derived from childhood glioblastoma multiforme (D-456 MG), childhood ependymoma (D-528 EP and D-612 EP), childhood medulloblastoma (D-425 MED), and adult malignant glioma (D-245 MG and D-54 MG), respectively, with corresponding tumor regressions in 10 of 10, 4 of 10, 8 of 10, 9 of 10, 9 of 10, and 10 of 10 treated mice, respectively. Delayed toxicity was seen more than 60 days after treatment, with 23 deaths in 100 treated animals, despite a median weight loss of only 0.06%. In mice bearing intracranial D-245 MG xenografts, treatment with VNP40101M at a dose of 18 mg/kg daily for five days produced a 50% increase in median survival compared with controls. Additional experiments conducted against subcutaneous D-245 MG xenografts by using reduced doses of 13.5 or 9.0 mg/kg daily for five days demonstrated tumor growth delays of 82.2 and 53.5 days, with corresponding tumor regressions in 8 of 9 and 9 of 10 treated mice, respectively (all values, p < 0.001), with one toxic death. These findings suggest that VNP40101M is active in the treatment of a wide range of human central nervous system tumors and warrants translation to the clinic.


Subject(s)
Brain Neoplasms/drug therapy , Hydrazines/therapeutic use , Neoplasms, Experimental/drug therapy , Prodrugs/therapeutic use , Sulfonamides/therapeutic use , Animals , Female , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Xenograft Model Antitumor Assays
9.
Cancer Chemother Pharmacol ; 60(1): 45-51, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17256135

ABSTRACT

Cloretazine (VNP40101M), a new sulfonylhydrazine alkylating agent, has demonstrated broad-spectrum anti-tumor activity in preclinical studies. In this study, Cloretazine was evaluated both as a monotherapy and in combination with fludarabine in murine tumor and human tumor xenograft models. Cloretazine significantly inhibited the growth of subcutaneously implanted tumors, including B16F10 murine melanoma in C57BL/6 mice, and H460 human lung carcinoma and WiDr human colon carcinoma in athymic nude CD1 mice. The inhibition of tumor growth by Cloretazine was dose dependent, increasing from 42.2 to 87% as the dose escalated from 100 to 150 mg/kg. Cloretazine showed equivalent efficacy but lower toxicity compared to cyclophosphamide in these models. The combination therapy, consisting of a single dose of 10 mg/kg Cloretazine plus five doses of 70 mg/kg fludarabine, given every other day intraperitoneally, significantly increased the long-term survival of BDF1 mice bearing the L1210 murine leukemia. On Day 65 post-tumor implantation, the combination therapy yielded a 90% survival rate compared to 40% for Cloretazine alone and 0% for fludarabine alone.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hydrazines/therapeutic use , Leukemia L1210/drug therapy , Melanoma, Experimental/drug therapy , Sulfonamides/therapeutic use , Xenograft Model Antitumor Assays/methods , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/chemistry , Cell Line, Tumor , Cyclophosphamide/administration & dosage , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor/methods , Humans , Hydrazines/administration & dosage , Hydrazines/chemistry , Injections, Intraperitoneal , Leukemia L1210/pathology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Nude , Sulfonamides/administration & dosage , Sulfonamides/chemistry , Survival Analysis , Time Factors , Tumor Burden/drug effects , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives , Weight Loss
10.
Science ; 313(5789): 936-40, 2006 Aug 18.
Article in English | MEDLINE | ID: mdl-16917054

ABSTRACT

NGC 6397 is the second closest globular star cluster to the Sun. Using 5 days of time on the Hubble Space Telescope, we have constructed an ultradeep color-magnitude diagram for this cluster. We see a clear truncation in each of its two major stellar sequences. Faint red main-sequence stars run out well above our observational limit and near to the theoretical prediction for the lowest mass stars capable of stable hydrogen burning in their cores. We also see a truncation in the number counts of faint blue stars, namely white dwarfs. This reflects the limit to which the bulk of the white dwarfs can cool over the lifetime of the cluster. There is also a turn toward bluer colors in the least luminous of these objects. This was predicted for the very coolest white dwarfs with hydrogen-rich atmospheres as the formation of H(2) and the resultant collision-induced absorption cause their atmospheres to become largely opaque to infrared radiation.

11.
Leuk Res ; 30(7): 813-22, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16478631

ABSTRACT

Triapine, an iron chelator and a potent inhibitor of ribonucleotide reductase, has significant anti-leukemia activity. A phase I study of Triapine in combination with ara-C was conducted in 32 patients with refractory acute leukemia and high-risk MDS. Triapine (105 mg/m2/day 6-h infusion) was followed immediately by ara-C [100 (n=4), 200 (n=6), 400 (n=7), or 800 (n=8)mg/m2/day] as an 18-h infusion for 5 consecutive days. Dose-limiting toxicities (DLTs) were observed at the 800 mg/m2 ara-C dose level (one patient each with grade 4 mucositis; grade 4 neutropenic colitis, sepsis; grade 4 neuropathy; and grade 4 hyperbilirubinemia). Therefore, the study was amended to include an ara-C dose level of 600 mg/m2/day, no DLTs occurred in seven patients treated at this dose level. Mean Triapine C(max) and AUC were 1.13 microg/mL and 251.5 minmicrog/mL. Of 31 evaluable patients, 4 (13%) (3 AML, 1 Ph+ALL) achieved a CR (1 at a dose of 800 mg/m2; 2 at 600 mg/m2; 1 at 200mg/m2). The recommended phase II regimen is Triapine 105 mg/m2/day followed by ara-C 600 mg/m2/day for 5 consecutive days every 3-6 weeks.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cytarabine/administration & dosage , Leukemia, Myeloid/drug therapy , Myelodysplastic Syndromes/drug therapy , Pyridines/administration & dosage , Thiosemicarbazones/administration & dosage , Acute Disease , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Cytarabine/adverse effects , Cytarabine/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Combinations , Female , Humans , Injections, Intravenous , Leukemia, Myeloid/diagnosis , Male , Maximum Tolerated Dose , Middle Aged , Myelodysplastic Syndromes/diagnosis , Pyridines/adverse effects , Pyridines/pharmacokinetics , Recurrence , Risk Factors , Thiosemicarbazones/adverse effects , Thiosemicarbazones/pharmacokinetics , Treatment Outcome
12.
Clin Cancer Res ; 11(13): 4827-34, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-16000580

ABSTRACT

PURPOSE: Genetically modified bacteria are a potentially powerful anticancer therapy due to their tumor targeting capacity, inherent antitumor activity, and ability to serve as efficient vectors for gene delivery. This study sought to characterize the acute and short-term toxicities and tumor colonization rates of a genetically modified Salmonella typhimurium (VNP20009) in dogs with spontaneous tumors, in the context of a phase I dose escalation trial. EXPERIMENTAL DESIGN: Forty-one pet dogs with a variety of malignant tumors received weekly or biweekly i.v. infusions of VNP20009, at doses ranging from 1.5 x 10(5) to 1 x 10(8) cfu/kg. Vital signs and clinicopathologic variables were monitored regularly. Incisional biopsies were obtained before and 1 week following the first infusion for histopathology and bacterial culture. RESULTS: The nominal maximum tolerated dose was 3 x 10(7) cfu/kg, with refractory fever and vomiting being the dose-limiting toxicities. One treatment-related acute death occurred. Bacteria were cultured from tumor tissue in 42% of cases. Thirty-five patients were evaluable for antitumor response. Major antitumor responses were seen in 15% (4 complete response and 2 partial response), and disease stabilization for at least 6 weeks in 10%. CONCLUSIONS: Administration of VNP20009 at doses with acceptable toxicity results in detectable bacterial colonization of tumor tissue and significant antitumor activity in tumor-bearing dogs.


Subject(s)
Neoplasms/drug therapy , Vaccines, Attenuated/therapeutic use , Animals , Bacterial Vaccines , Diarrhea/chemically induced , Dog Diseases/drug therapy , Dog Diseases/immunology , Dogs , Dose-Response Relationship, Drug , Female , Fever/chemically induced , Male , Neoplasms/pathology , Neoplasms/veterinary , Salmonella typhimurium/immunology , Treatment Outcome , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology , Vomiting/chemically induced
14.
Hum Gene Ther ; 13(10): 1225-33, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12133275

ABSTRACT

The study was designed to evaluate whether TAPET-CD, an attenuated strain of Salmonella typhimurium expressing Escherichia coli cytosine deaminase (CD), was capable of converting nontoxic 5-fluorocytosine (5-FC) to the active antitumor agent 5-fluorouracil (5-FU). The antitumor effect of TAPET-CD plus 5-FC against subcutaneously implanted colon tumors was also evaluated. TAPET-CD was given to tumor-bearing mice by a single bolus intravenous administration followed with 5-FC by intraperitoneal administration. TAPET-CD accumulated in tumors at levels 1000-fold higher than that in normal tissues and high levels of 5-FU were detected in tumors in mice treated with both TAPET-CD and 5-FC. No 5-FU could be detected in normal tissues. Inhibition of tumor growth was observed in mice treated with either TAPET-CD alone or TAPET-CD in combination with 5-FC (TAPET-CD/5-FC), but not with 5-FC alone. TAPET-CD/5-FC inhibited tumor growth by 88%-96%, compared to TAPET-CD alone, which inhibited tumor growth by 38%-79%. These data suggest that tumor-targeting Salmonella could be used to deliver prodrug-converting enzyme selectively to tumors and produced anti-tumor effects when the corresponding prodrug was also given.


Subject(s)
Antineoplastic Agents/pharmacology , Fluorouracil/therapeutic use , Nucleoside Deaminases/pharmacology , Salmonella typhimurium/physiology , Animals , Biotransformation , Colonic Neoplasms/therapy , Cytosine Deaminase , Escherichia coli/enzymology , Female , Flucytosine/metabolism , Fluorouracil/administration & dosage , Fluorouracil/metabolism , Genetic Markers , Humans , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Nucleoside Deaminases/genetics , Organ Specificity , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Tumor Cells, Cultured
15.
Curr Opin Drug Discov Devel ; 5(2): 194-9, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11926125

ABSTRACT

The development of novel cancer therapies that are selective for cancer cells with limited toxicity to normal tissues is a challenge for oncology researchers. Microorganisms, such as viruses with selectivity for tumor cells or tumor micro-environments, have been investigated as potential arsenals for decades. Genetically-modified, non-pathogenic bacteria have begun to emerge as potential antitumor agents, either to provide direct tumoricidal effects or to deliver tumoricidal molecules. Attenuated Salmonella, Clostridium and Bifidobacterium are capable of multiplying selectively in tumors and inhibiting their growth, representing a new approach for cancer treatment. Because of their selectivity for tumor tissues, these bacteria would also be ideal vectors for delivering therapeutic proteins to tumors. VNP20009, an attenuated strain of Salmonella typhimurium, and its derivative, TAPET-CD, which expresses an Escherichia coli cytosine deaminase (CD), are particularly promising, and are currently undergoing phase I clinical trials in cancer patients.


Subject(s)
Bacterial Physiological Phenomena , Drug Delivery Systems , Neoplasms/therapy , Proteins/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Bifidobacterium/metabolism , Clostridium/metabolism , Humans , Salmonella/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...