Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
1.
Org Biomol Chem ; 14(3): 963-9, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26620576

ABSTRACT

The one-pot borylation/Suzuki reaction is a very efficient means of accessing cross-coupling products of two aryl-halide partners that generally requires the use of specific catalysts or ligands and/or relatively long reaction times. This new microwave-assisted method provides a quick one-pot borylation/Suzuki reaction protocol that we applied to the synthesis of various bi- or poly-aryl scaffolds, including a variety of aryl and heteroaryl ring systems and the core frameworks of kinase inhibitors vemurafenib and GDC-0879.


Subject(s)
Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Catalysis , Dose-Response Relationship, Drug , Indenes/chemistry , Indoles/chemistry , Microwaves , Molecular Structure , Organometallic Compounds/chemistry , Palladium/chemistry , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Structure-Activity Relationship , Sulfonamides/chemistry , Vemurafenib
2.
J Med Chem ; 47(10): 2651-8, 2004 May 06.
Article in English | MEDLINE | ID: mdl-15115406

ABSTRACT

Nine new nitrogen mustard compounds derived from 2,6-difluoro-4-hydroxy- (3a-e) and 2,6-difluoro-4-amino- (4a-d) aniline were synthesized as potential prodrugs. They were designed to be activated to their corresponding 3,5-difluorophenol and -aniline (4)-nitrogen mustards by the enzyme carboxypeptidase G2 (CPG2) in gene-directed enzyme prodrug therapy (GDEPT) models. The compounds were tested for cytotoxicity in the MDA MB-361 breast adenocarcinoma. The cell line was engineered to express stably either CPG2 tethered to the cell surface stCPG2-(Q)3 or beta-galactosidase (beta-Gal) as control. The cytotoxicity differentials were calculated between CPG 2-expressing and -nonexpressing cells and yielded different results for the two series of prodrugs despite their structural similarities. While the phenol compounds are ineffective as prodrugs, their aniline counterparts exhibit outstanding activity in the tumor cell lines expressing CPG2. [3,5-Difluoro-4-[bis(2-chloroethyl)amino]phenyl]carbamoyl-l-glutamic acid gave a differential of >227 in MDA MB361 cells as compared with 19 exhibited by 4-[(2-chloroethyl)(2-mesyloxyethyl)amino]benzoyl-l-glutamic acid, 1a, which has been in clinical trials.


Subject(s)
Antineoplastic Agents/metabolism , Glutamic Acid/metabolism , Nitrogen Mustard Compounds/metabolism , Prodrugs/metabolism , gamma-Glutamyl Hydrolase/metabolism , Aniline Mustard/analogs & derivatives , Aniline Mustard/chemical synthesis , Aniline Mustard/metabolism , Aniline Mustard/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Benzene Derivatives/chemical synthesis , Benzene Derivatives/metabolism , Benzene Derivatives/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Genetic Therapy , Glutamic Acid/analogs & derivatives , Glutamic Acid/chemical synthesis , Glutamic Acid/pharmacology , Half-Life , Humans , Mice , Neoplasm Transplantation , Nitrogen Mustard Compounds/chemical synthesis , Nitrogen Mustard Compounds/pharmacology , Prodrugs/chemical synthesis , Prodrugs/pharmacology , Structure-Activity Relationship , Transplantation, Heterologous , gamma-Glutamyl Hydrolase/chemistry , gamma-Glutamyl Hydrolase/genetics
3.
Br J Cancer ; 88(10): 1622-30, 2003 May 19.
Article in English | MEDLINE | ID: mdl-12771932

ABSTRACT

We have generated fusion proteins between vascular endothelial growth factor (VEGF) and the bacterial enzyme carboxypeptidase G2 (CPG2) that can activate the prodrug 4-[(2-chloroethyl)(2-mesyloxyethyl)amino]benzoyl-L-glutamic acid (CMDA). Three asparagine residues of CPG2 were mutated to glutamine (CPG2(Q)3) to prevent glycosylation during secretion, and truncations of VEGF(165) were fused to either the C- or N-terminal of CPG2. The K(m) of the fusion proteins (37.5 microM) was similar to that of secreted CPG2(Q)3 (29.5 microM) but greater than that of wild-type CPG2 (8 microM). The affinity of the fusion proteins for VEGF receptor-2 (VEGFR2) (K(d)=0.5-1.1 nM) was similar to that of [(125)I]VEGF (K(d)=0.5 nM) (ELISA) or slightly higher (K(d)=1.3-9.6 nM) (competitive RIA). One protein, VEGF(115)-CPG2(Q)3-H(6), possessed 140% of the enzymic activity of secreted CPG2(Q)3, and had a faster half-maximal binding time for VEGFR2 (77 s), than the other candidates (330 s). In vitro, VEGF(115)-CPG2(Q)3-H(6) targeted CMDA cytotoxicity only towards VEGFR-expressing cells. The plasma half-life of VEGF(115)-CPG2(Q)3-H(6) in vivo was 3 h, comparable to equivalent values observed in ADEPT. We conclude that enzyme prodrug therapy using VEGF as a targeting moiety represents a promising novel antitumour therapy, with VEGF(115)-CPG2(Q)3-H(6) being a lead candidate.


Subject(s)
Endothelial Growth Factors/pharmacology , Glutamates/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Lymphokines/pharmacology , Nitrogen Mustard Compounds/pharmacology , Prodrugs/pharmacology , Vascular Endothelial Growth Factor Receptor-2/genetics , gamma-Glutamyl Hydrolase/pharmacology , Adenocarcinoma/pathology , Endothelial Growth Factors/genetics , Endothelium/cytology , Female , Glutamine , Humans , Intercellular Signaling Peptides and Proteins/genetics , Lymphokines/genetics , Mutagenesis, Site-Directed , Neovascularization, Pathologic , Ovarian Neoplasms/pathology , Plasmids , Point Mutation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-2/drug effects , Vascular Endothelial Growth Factors , gamma-Glutamyl Hydrolase/genetics
4.
Adv Drug Deliv Rev ; 53(3): 247-64, 2001 Dec 31.
Article in English | MEDLINE | ID: mdl-11744170

ABSTRACT

A great deal of interest has surrounded the activities of monoclonal antibodies (mAbs), and mAb-drug, toxin and radionuclide conjugates for the treatment of human cancers. In the last few years, a number of new mAb-based reagents have been clinically approved (Rituxan, Herceptin, and Panorex), and several others are now in advanced clinical trials. Successful therapeutic treatment of solid tumors with drug conjugates of such macromolecules must overcome the barriers to penetration within tumor masses, antigen heterogeneity, conjugated drug potency, and efficient drug release from the mAbs inside tumor cells. An alternative strategy for drug delivery involves a two-step approach to cancer therapy in which mAbs are used to localize enzymes to tumor cell surface antigens. Once the conjugate binds to the cancer cells and clears from the systemic circulation, antitumor prodrugs are administered that are catalytically converted to active drugs by the targeted enzyme. The drugs thus released are capable of penetrating within the tumor mass and eliminating both cells that have and have not bound the mAb-enzyme conjugate. Significant therapeutic effects have been obtained using a broad range of enzymes along with prodrugs that are derived from both approved anticancer drugs and highly potent experimental agents. This review focuses on the activities of several mAb-enzyme/prodrug combinations, with an emphasis on those that have provided mechanistic insight, clinical activity, novel protein constructs, and the potential for reduced immunogenicity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Enzymes/pharmacology , Prodrugs/pharmacology , Animals , Antibodies, Monoclonal/chemistry , Antineoplastic Agents/metabolism , Biotransformation , Enzymes/chemistry , Humans , Prodrugs/metabolism
5.
Int J Cancer ; 93(1): 123-30, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11391631

ABSTRACT

Escherichia coli B nitroreductase (NR) has been expressed stably in MDA-MB-361 human breast adenocarcinoma cells either as the wild-type protein (wtNR), which is distributed evenly between the cytoplasmic and nuclear compartments, or targeted to the mitochondrion (mtNR). Whereas bacterial NR is active as a dimer, a proportion of wtNR is monomeric. In contrast, mtNR is mostly dimeric, suggesting that it adopts a more stable, native conformation. Despite this, when tested in gene-directed enzyme prodrug therapy cell cytotoxicity studies, cells expressing wtNR or mtNR had similar sensitivity to the prodrug CB1954 and mounted similar bystander killing effects. Furthermore, when short prodrug exposures were given, wtNR was more efficient at killing cells than mtNR. These data demonstrate that the site of enzyme expression and prodrug activation is an important variable that requires consideration in suicide gene therapy approaches.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Aziridines/pharmacokinetics , Genetic Therapy , Nitroreductases/metabolism , Prodrugs/pharmacokinetics , Adenocarcinoma , Antineoplastic Agents/toxicity , Aziridines/toxicity , Base Sequence , Breast Neoplasms , Cell Survival/drug effects , Cloning, Molecular , Dimerization , Enzyme Activation , Escherichia coli/enzymology , Female , Humans , Molecular Sequence Data , Nitroreductases/genetics , Prodrugs/toxicity , Recombinant Proteins/metabolism , Restriction Mapping , Transfection , Tumor Cells, Cultured
6.
Cancer Gene Ther ; 7(10): 1348-56, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11059693

ABSTRACT

The bacterial enzyme carboxypeptidase G2 (CPG2) can be expressed both intracellularly (CPG2*) or tethered to the outer surface (stCPG2(Q)3) of mammalian cells, where it is able to activate mustard prodrugs for use in suicide gene therapy protocols. Here we compare the properties of CPG2 expressed in these two locations. CPG2 is active as a dimer, and one of the mutations required to block glycosylation of stCPG2(Q)3 destabilizes the dimers. Some of the mutations to this site partially correct the dimerization defect and recover a proportion of the activity. Surface tethering also recovers some enzyme activity, but through an unknown mechanism. The efficacy of CPG2 in these two locations is compared with the tumor cell lines A2780, SK-OV-3, and WiDr, which are sensitized to the prodrug 4-([2-chloroethyl][2-mesyloxyethyl]amino)benzoyl-L-glutamic acid (CMDA) by both CPG2* and stCPG2(Q)3 expression in suicide gene therapy protocols in vitro. We find that stCPG2(Q)3 is a more efficient mediator of CMDA-dependent cell killing than CPG2*. Lower levels of stCPG2(Q)3 activity are required to give cell killing that can only be achieved by higher levels of CPG2*. In bystander effect assays, low levels of stCPG2(Q)3 are required for efficient killing, whereas relatively high levels of CPG2* activity are required. Also, shorter exposures to prodrug are required for cell killing when stCPG2(Q)3 is expressed compared with when CPG2* is expressed. These data demonstrate that the location of the enzyme in the cell is more important than the enzyme activity as the determinant in mediating cytotoxicity.


Subject(s)
Colorectal Neoplasms/drug therapy , Genetic Therapy/methods , Glutamates/pharmacology , Nitrogen Mustard Compounds/pharmacology , Ovarian Neoplasms/drug therapy , Prodrugs/pharmacology , gamma-Glutamyl Hydrolase/metabolism , Animals , COS Cells , Chlorocebus aethiops , Colorectal Neoplasms/enzymology , DNA Primers/chemistry , Electrophoresis, Polyacrylamide Gel , Female , Genetic Vectors , Humans , Mutation , Ovarian Neoplasms/enzymology , Polymerase Chain Reaction , Tumor Cells, Cultured , gamma-Glutamyl Hydrolase/genetics
9.
Clin Cancer Res ; 6(3): 765-72, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10741695

ABSTRACT

In antibody-directed enzyme prodrug therapy, an enzyme conjugated to an antitumor antibody is given i.v. and localizes in the tumor. A prodrug is then given, which is converted to a cytotoxic drug selectively in the tumor. Ten patients with colorectal carcinoma expressing carcinoembryonic antigen received antibody-directed enzyme prodrug therapy with A5B7 F(ab')2 antibody to carcinoembryonic antigen conjugated to carboxypeptidase G2 (CPG2). A galactosylated antibody directed against the active site of CPG2 (SB43-gal) was given to clear and inactivate circulating enzyme. A benzoic acid mustard-glutamate prodrug was given when plasma enzyme levels had fallen to a predetermined safe level, and this was converted by CPG2 in the tumor into a cytotoxic form. Enzyme levels derived from quantitative gamma camera imaging and from direct measurements in plasma and tumor biopsies showed that the median tumor:plasma ratio of enzyme exceeded 10000:1 at the time of prodrug administration. Enzyme concentrations in the tumor (median, 0.47 units g(-1)) were sufficient to generate cytotoxic levels of active drug. The concentration of prodrug needed for optimal conversion (Km) of 3 microM was achieved. Prodrug conversion to drug was shown by finding detectable levels of drug in plasma. There was evidence of tumor response; one patient had a partial response, and six patients had stable disease for a median of 4 months after previous tumor progression (one of these six had a tumor marker response). Manageable neutropenia and thrombocytopenia occurred. Conditions for effective antitumor therapy were met, and there was evidence of tumor response in colorectal cancer.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Colorectal Neoplasms/therapy , Glutamates/therapeutic use , Nitrogen Mustard Compounds/therapeutic use , Prodrugs/therapeutic use , gamma-Glutamyl Hydrolase/administration & dosage , Adult , Aged , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/chemistry , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Carcinoembryonic Antigen/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Glutamates/adverse effects , Glutamates/pharmacokinetics , Humans , Male , Middle Aged , Neutropenia/chemically induced , Nitrogen Mustard Compounds/adverse effects , Nitrogen Mustard Compounds/pharmacokinetics , Prodrugs/adverse effects , Prodrugs/pharmacokinetics , Survival Analysis , Thrombocytopenia/chemically induced , Treatment Outcome , gamma-Glutamyl Hydrolase/blood , gamma-Glutamyl Hydrolase/chemistry
10.
Hum Gene Ther ; 11(2): 285-92, 2000 Jan 20.
Article in English | MEDLINE | ID: mdl-10680842

ABSTRACT

The role of the bystander effect in the treatment of a human breast carcinoma xenograft was studied by suicide gene therapy with carboxypeptidase G2 (CPG2) and CMDA. Cells expressing enzymatically active surface-tethered bacterial CPG2 [stCPG2(Q)3] were mixed with control beta-galactosidase (beta-Gal)-expressing cells to give stCPG2(Q)3:beta-Gal ratios of, respectively: group 1, 0:100; group 2, 10:90; group 3, 50:50; and group 4, 100:0. Four days after injection of the cells into nude mice, the prodrug 4-[(2-chloroethyl)(2-mesyloxyethyl)amino]benzoyl-L-glutamic acid (CMDA) was administered. Tumor growth delay correlated well with the levels of stCPG2(Q)3 expression: group 1, 0 day delay; group 2, 10 days; group 3, 16 days; and group 4, 90 days. Similarly, the number of cures was strongly correlated to the levels of stCPG2(Q)3 activity: group 1, zero of six cured; group 2, one of six cured; group 3, three of six cured and group 4, four of six cured. There was a good correlation between CPG2 enzyme activity in the tumors and the number of cures. The majority of cells from groups 2 and 3 were apoptotic whereas those from group 1 were not, indicating a substantial bystander effect in the tumors. These results suggest that a bystander effect plays a major role in suicide gene therapy regimens with stCPG2(Q)3 and CMDA.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Genetic Therapy/methods , Glutamates/pharmacology , Nitrogen Mustard Compounds/pharmacology , Prodrugs/pharmacology , gamma-Glutamyl Hydrolase/genetics , Animals , Apoptosis/drug effects , Body Weight/drug effects , Breast Neoplasms/drug therapy , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Time Factors
11.
Cancer Res ; 59(16): 3998-4003, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10463598

ABSTRACT

The irregular nature of solid tumor vasculature produces a heterogeneous distribution of antibody-targeted therapies within the tumor mass, which frequently results in reduced therapeutic efficacy. We have, therefore, combined two complementary therapies: Antibody-directed Enzyme Prodrug Therapy (ADEPT), which targets tumor cells, and an agent that selectively destroys tumor vasculature. A single i.p. dose (27.5 mg/kg) of the drug 5,6-dimethylxanthenone-4-acetic acid (DMXAA), given to nude mice bearing the LS174T colorectal xenograft, destroyed all but a peripheral rim of tumor cells, without enhancing survival. The ADEPT system, in which a pretargeted enzyme activates a prodrug, consisted of the F(ab')2 fragment of anti-carcinoembryonic antigen antibody A5B7 conjugated to the bacterial enzyme carboxypeptidase G2 and the prodrug 4-[(2-chloroethyl)(2-mesyloxyethyl)amino]benzoyl-L-glutamic acid, which was given i.p. in three doses of 500 mg/kg at 72, 84, and 96 h post-conjugate administration (25 units of carboxypeptidase G2). The antibody-enzyme conjugate could be selectively retained at approximately twice the control levels by administration of the antivascular agent at the time of optimal conjugate localization within the tumor (20 h post-conjugate administration), as demonstrated by gamma counting, phosphor plate image analysis, and active enzyme measurement. This resulted in significantly enhanced tumor growth inhibition in groups of six mice, compared to conventional ADEPT therapy, with no concomitant increase in systemic toxicity. In a separate experiment, aimed at trapping the prodrug within the tumor, a 16-fold increase over control values was produced (means, 44.8 versus 2.8 microg/g tumor) when DMXAA was given 4 h prior to 4-[(2-chloroethyl)(2-mesyloxyethyl)amino]benzoyl-L-glutamic acid. The therapeutic window was small, with no significant enhancement of prodrug retention when DMXAA was given at either earlier or later time points. This correlated with the time of vascular shut-down induced by the antivascular agent. We are currently investigating whether it is more advantageous to trap increased levels of conjugate or prodrug within the tumor for maximal enhancement of conventional ADEPT. These studies demonstrate that combined use of antibody-directed and antivascular therapies can significantly benefit the therapeutic outcome of either strategy alone.


Subject(s)
Antineoplastic Agents/administration & dosage , Colorectal Neoplasms/drug therapy , Glutamates/administration & dosage , Immunoconjugates/administration & dosage , Neovascularization, Pathologic/drug therapy , Nitrogen Mustard Compounds/administration & dosage , Prodrugs/administration & dosage , Xanthenes/administration & dosage , Xanthones , Animals , Carcinoembryonic Antigen/immunology , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/immunology , Female , Humans , Immunoglobulin Fab Fragments/administration & dosage , Immunoglobulin Fab Fragments/immunology , Mice , Mice, Nude , Neoplasm Transplantation , Transplantation, Heterologous , gamma-Glutamyl Hydrolase/administration & dosage
12.
J Med Chem ; 42(13): 2485-9, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10395490

ABSTRACT

Four novel potential prodrugs derived from daunorubicin (8, 10) and doxorubicin (12, 14) were designed and synthesized. They are self-immolative prodrugs for suicide gene therapy activation by the enzyme carboxypeptidase G2 (CPG2) subsequently releasing the corresponding anthracyclines, by a 1,6-elimination mechanism. A mammary carcinoma cell line (MDA MB 361) was engineered to express CPG2 intracellularly (CPG2) or extracellularly, tethered to the outer cell membrane (stCPG2(Q)3). The prodrugs derived from doxorubicin showed prodrug/drug cytotoxicity differentials of 21-fold (compound 12) and 23-fold (compound 14). Prodrug 12 underwent an 11-fold activation when assayed in the cell line expressing externally surface-tethered CPG2.


Subject(s)
Antineoplastic Agents/chemical synthesis , Daunorubicin/chemistry , Doxorubicin/chemistry , Prodrugs/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Membrane/enzymology , Daunorubicin/metabolism , Doxorubicin/metabolism , Genetic Therapy , Humans , Prodrugs/pharmacology , Tumor Cells, Cultured , gamma-Glutamyl Hydrolase/chemistry , gamma-Glutamyl Hydrolase/metabolism
13.
EMBO J ; 18(8): 2137-48, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10205168

ABSTRACT

The Raf family of serine/threonine protein kinases couple growth factor receptor stimulation to mitogen activated protein kinase activation, but their own regulation is poorly understood. Using phospho-specific antisera, we show that activated Raf-1 is phosphorylated on S338 and Y341. Expression of Raf-1 with oncogenic Ras gives predominantly S338 phosphorylation, whereas activated Src gives predominantly Y341 phosphorylation. Phosphorylation at both sites is maximal only when both oncogenic Ras and activated Src are present. Raf-1 that cannot interact with Ras-GTP is not phosphorylated, showing that phosphorylation is Ras dependent, presumably occurring at the plasma membrane. Mutations which prevent phosphorylation at either site block Raf-1 activation and maximal activity is seen only when both are phosphorylated. Mutations at S339 or Y340 do not block Raf-1 activation. While B-Raf lacks a tyrosine phosphorylation site equivalent to Y341 of Raf-1, S445 of B-Raf is equivalent to S338 of Raf-1. Phosphorylation of S445 is constitutive and is not stimulated by oncogenic Ras. However, S445 phosphorylation still contributes to B-Raf activation by elevating basal and consequently Ras-stimulated activity. Thus, there are considerable differences between the activation of the Raf proteins; Ras-GTP mediates two phosphorylation events required for Raf-1 activation but does not regulate such events for B-Raf.


Subject(s)
Proto-Oncogene Proteins c-raf/metabolism , Serine/metabolism , Tyrosine/metabolism , Animals , COS Cells , Cell Membrane/metabolism , Epidermal Growth Factor/pharmacology , Phosphorylation , Tetradecanoylphorbol Acetate/pharmacology
14.
Anticancer Drug Des ; 14(6): 517-38, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10834273

ABSTRACT

Antibody- and gene-directed enzyme prodrug therapy are two-step targeting strategies designed to improve the selectivity of antitumour agents. The approaches are based on the activation of specially designed prodrugs by antibody-enzyme conjugates targeted to tumour-associated antigens (ADEPT) or by enzymes expressed by exogenous genes in tumour cells (GDEPT). Herein the design, synthesis, physico-chemical and biological properties, kinetics and clinical trials of the prodrugs and the enzymes carboxypeptidase G2 and nitroreductase are reviewed for ADEPT and GDEPT.


Subject(s)
Genetic Therapy/methods , Immunotherapy/methods , Neoplasms/therapy , Prodrugs , Animals , Antibodies, Neoplasm/immunology , Drug Design , Humans
15.
Curr Opin Mol Ther ; 1(4): 480-6, 1999 Aug.
Article in English | MEDLINE | ID: mdl-11713763

ABSTRACT

Gene-directed enzyme prodrug therapy (GDEPT) is a promising two-step treatment for solid malignant tumors. In the first step, the gene for a foreign enzyme is administered and directed to the tumor, where it may be expressed using specific transcriptional elements. In the second step, prodrugs are administered and activated by the foreign enzyme expressed at the tumor. This review focuses on the progress from the end of 1997 to date. Important issues, such as viral and non-viral vectors, new enzyme/prodrug systems, new strategies, advances in the understanding of the bystander effects, the comparison of different systems used in GDEPT and clinical trials are outlined.


Subject(s)
Enzymes/genetics , Genetic Therapy , Neoplasms/drug therapy , Prodrugs/pharmacokinetics , Adenoviridae/genetics , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Biotransformation , Bystander Effect , Clinical Trials as Topic , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Cytosine Deaminase , Dependovirus/genetics , Drug Carriers , Enzyme Activation , Enzyme Induction , Enzymes/metabolism , Fluorouracil/pharmacokinetics , Fluorouracil/therapeutic use , Forecasting , Genetic Vectors/administration & dosage , Genetic Vectors/therapeutic use , Liposomes , Neoplasms/genetics , Nucleoside Deaminases/genetics , Nucleoside Deaminases/metabolism , Rabbits , Rats , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid , Retroviridae/genetics , Simplexvirus/genetics , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Transcription, Genetic , Viral Proteins/genetics , Viral Proteins/metabolism , gamma-Glutamyl Hydrolase/genetics , gamma-Glutamyl Hydrolase/metabolism
16.
J Med Chem ; 41(26): 5297-309, 1998 Dec 17.
Article in English | MEDLINE | ID: mdl-9857097

ABSTRACT

Four new potential self-immolative prodrugs derived from phenol and aniline nitrogen mustards, four model compounds derived from their corresponding fluoroethyl analogues and two new self-immolative linkers were designed and synthesized for use in the suicide gene therapy termed GDEPT (gene-directed enzyme prodrug therapy). The self-immolative prodrugs were designed to be activated by the enzyme carboxypeptidase G2 (CPG2) releasing an active drug by a 1, 6-elimination mechanism via an unstable intermediate. Thus, N-[(4-¿[4-(bis¿2-chloroethyl¿amino)phenoxycarbonyloxy]methyl¿pheny l)c arbamoyl]-L-glutamic acid (23), N-[(4-¿[4-(bis¿2-chloroethyl¿amino)phenoxycarbonyloxy]methyl¿pheno xy) carbonyl]-L-glutamic acid (30), N-[(4-¿[N-(4-¿bis[2-chloroethyl]amino¿phenyl)carbamoyloxy]methyl¿+ ++phen oxy)carbonyl]-L-glutamic acid (37), and N-[(4-¿[N-(4-¿bis[2-chloroethyl]amino¿phenyl)carbamoyloxy]methyl¿+ ++phen yl)carbamoyl]-L-glutamic acid (40) were synthesized. They are bifunctional alkylating agents in which the activating effects of the phenolic hydroxyl or amino functions are masked through an oxycarbonyl or a carbamoyl bond to a benzylic spacer which is itself linked to a glutamic acid by an oxycarbonyl or a carbamoyl bond. The corresponding fluoroethyl compounds 25, 32, 42, and 44 were also synthesized. The rationale was to obtain model compounds with greatly reduced alkylating abilities that would be much less reactive with nucleophiles compared to the corresponding chloroethyl derivatives. This enabled studies of these model compounds as substrates for CPG2, without incurring the rapid and complicated decomposition pathways of the chloroethyl derivatives. The prodrugs were designed to be activated to their corresponding phenol and aniline nitrogen mustard drugs by CPG2 for use in GDEPT. The synthesis of the analogous novel parent drugs (21b, 51) is also described. A colorectal cell line was engineered to express CPG2 tethered to the outer cell surface. The phenylenediamine compounds were found to behave as prodrugs, yielding IC50 prodrug/IC50 drug ratios between 20- and 33-fold (for 37 and 40) and differentials of 12-14-fold between CPG2-expressing and control LacZ-expressing clones. The drugs released are up to 70-fold more potent than 4-[(2-chloroethyl)(2-mesyloxyethyl)amino]benzoic acid that results from the prodrug 4-[(2-chloroethyl)(2-mesyloxyethyl)amino]benzoyl-L-glutamic acid (CMDA) which has been used previously for GDEPT. These data demonstrate the viability of this strategy and indicate that self-immolative prodrugs can be synthesized to release potent mustard drugs selectively by cells expressing CPG2 tethered to the cell surface in GDEPT.


Subject(s)
Antineoplastic Agents, Alkylating/chemical synthesis , Genetic Therapy , Mechlorethamine/analogs & derivatives , Mechlorethamine/chemical synthesis , Prodrugs/chemical synthesis , Animals , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/pharmacokinetics , Antineoplastic Agents, Alkylating/pharmacology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Screening Assays, Antitumor , Inhibitory Concentration 50 , Mechlorethamine/metabolism , Mechlorethamine/pharmacology , Mice , Mutation , Prodrugs/metabolism , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Transfection , Tumor Cells, Cultured , gamma-Glutamyl Hydrolase/biosynthesis , gamma-Glutamyl Hydrolase/genetics , gamma-Glutamyl Hydrolase/metabolism
19.
Cancer Gene Ther ; 4(4): 229-38, 1997.
Article in English | MEDLINE | ID: mdl-9253508

ABSTRACT

Expression of genes encoding prodrug-activating enzymes can increase the susceptibility of tumor cells to prodrugs, and may ultimately achieve a better therapeutic index than conventional chemotherapy. CB1954 is a weak, monofunctional alkylating agent which can be activated by Escherichia coli nitroreductase to a potent dysfunctional alkylating agent which crosslinks DNA. We have inserted the nitroreductase gene into an LNCX-based retroviral vector, to allow efficient gene transfer and expression in colorectal (LS174T) and pancreatic (SUIT2, BxPC3, and AsPC1) cancer cell lines. A clone of LS174T cells expressing nitroreductase showed > 50-fold increased sensitivity to CB1954, and nitroreductase-expressing clones of pancreatic tumor lines were up to approximately 500-fold (SUIT2) more sensitive than parental cells. Concentrations of CB1954 minimally toxic to nontransduced cells achieved 100% cell death in a 50:50 mix of parental cells with SUIT2 cells expressing nitroreductase; and marked "bystander" cell killing was seen with just 10% of cells expressing nitroreductase. Significant bystander cell killing was dependent on a high cell density. In conjunction with regional delivery of vectors and tumor selectivity of cell entry and/or gene expression, nitroreductase and CB1954 may be an attractive combination for prodrug-activating enzyme gene therapy of colorectal and pancreatic cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Aziridines/therapeutic use , Colorectal Neoplasms/therapy , Genetic Therapy , Nitroreductases/genetics , Pancreatic Neoplasms/therapy , Prodrugs/therapeutic use , Antineoplastic Agents/pharmacology , Aziridines/pharmacology , Dose-Response Relationship, Drug , Escherichia coli/genetics , Genetic Vectors , Humans , Prodrugs/pharmacology , Retroviridae , Tumor Cells, Cultured
20.
Expert Opin Investig Drugs ; 6(6): 685-703, 1997 Jun.
Article in English | MEDLINE | ID: mdl-15989636

ABSTRACT

Gene-directed enzyme prodrug therapy (GDEPT) is a promising, new, two-step treatment for cancer chemotherapy. In the first step, the gene for a foreign enzyme is administered and is directed to the tumour, where it is expressed by the use of specific promoters. In the second step, injected prodrugs are activated by the foreign enzyme. The design and synthesis of prodrugs able to undergo enzymatic activation in such systems is an essential component. This review focuses on the requirements which must be fulfilled by the components of GDEPT systems in order for this therapy to be considered a realistic possibility. A special emphasis is placed on the description of the prodrugs used in GDEPT protocols and the requirement for a bystander effect is also discussed.

SELECTION OF CITATIONS
SEARCH DETAIL
...