Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Cereb Cortex ; 29(1): 1-16, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29136113

ABSTRACT

Piriform cortex (PC) is a 3-layer paleocortex receiving primary afferent input from the olfactory bulb. The past decade has seen significant progress in understanding the synaptic, cellular and functional organization of PC, but PC embryogenesis continues to be enigmatic. Here, using birthdating strategies and clonal analyses, we probed the early development and laminar specificity of neurogenesis/gliogenesis as it relates to the organization of the PC. Our data demonstrate a temporal sequence of laminar-specific neurogenesis following the canonical "inside-out" pattern, with the notable exception of PC Layer II which exhibited an inverse "outside-in" temporal neurogenic pattern. Of interest, we found no evidence of a neurogenic gradient along the anterior to posterior axis, although the timing of neuronal migration and laminar development was delayed rostrally by approximately 24 h. To begin probing if lineage affected cell fate in the PC, we labeled PC neuroblasts using a multicolor technique and analyzed their laminar organization. Our results suggested that PC progenitors were phenotypically committed to reach specific layers early in the development. Collectively, these studies shed new light on the determinants of the laminar specificity of neuronal/glial organization in PC and the likely role of subpopulations of committed progenitors in regulating PC embryogenesis.


Subject(s)
Cell Lineage/physiology , Cell Movement/physiology , Neurogenesis/physiology , Neuroglia/physiology , Piriform Cortex/cytology , Piriform Cortex/growth & development , Animals , Female , HEK293 Cells , Humans , Male , Mice , Pregnancy
2.
Anal Biochem ; 508: 34-7, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27188264

ABSTRACT

Alkylating agents are a significant class of environmental carcinogens as well as commonly used anticancer therapeutics. Traditional alkylating activity assays have utilized the colorimetric reagent 4-(4-nitrobenzyl)pyridine (4NBP). However, 4NBP based assays have a relatively low sensitivity towards harder, more oxophilic alkylating species and are not well suited for the identification of the trapped alkyl moiety due to adduct instability. Herein we describe a method using water as the trapping agent which permits the trapping of simple alkylating electrophiles with a comparatively wide range of softness/hardness and permits the identification of donated simple alkyl moieties.


Subject(s)
Alcohols/chemistry , Alkylating Agents/analysis , Alkylating Agents/isolation & purification , Carcinogens, Environmental/analysis , Carcinogens, Environmental/isolation & purification , Chemistry Techniques, Analytical/methods , Chemistry Techniques, Analytical/standards , Fresh Water/chemistry
3.
J Transl Sci ; 2(2): 117-124, 2016.
Article in English | MEDLINE | ID: mdl-27076919

ABSTRACT

Although epithelial ovarian cancers (EOCs) are initially treated with platinum-based chemotherapy, EOCs vary in platinum responsiveness. Cataloging antineoplastic agents according to their effectiveness against platinum-resistant and platinum-sensitive EOC cell lines is valuable for development of therapeutic strategies to avoid platinum inefficacy and to exploit platinum sensitivity. TOV-21G devoid of FANCF expression, OV-90 and SKOV-3 were employed as examples of platinum-sensitive, platinum-intermediate and platinum-resistant cell lines, respectively. Antineoplastic agents examined included mitomycin C, doxorubicin, etoposide, gemcitabine, chlorambucil, paclitaxel, triapine and X-rays. Their effectiveness against cell lines was analyzed by clonogenic assays. Cytotoxic profiles of mitomycin C and carboplatin were similar, with mitomycin C exhibiting greater potency and selectivity against TOV-21G than carboplatin. Cytotoxic profiles of doxorubicin, etoposide and X-rays overlapped with that of carboplatin, while OV-90 overexpressing Rad51 was more resistant to chlorambucil than SKOV-3. The efficacy of paclitaxel and triapine was independent of platinum sensitivity or resistance. Consistent with these cytotoxic profiles, cisplatin/mitomycin C, triapine, and paclitaxel differed in the capacity to induce phosphorylation of H2AX, and produced unique inhibitory patterns of DNA/RNA syntheses in HL-60 human leukemia cells. Paclitaxel and triapine in combination produced additive antitumor effects in M109 murine lung carcinoma. In conclusion, mitomycin C is potentially more effective against Fanconi anemia pathway-deficient EOCs than carboplatin. Doxorubicin and etoposide, because of their overlapping cytotoxic properties with carboplatin, are unlikely to be efficacious against platinum-refractory EOCs. Paclitaxel and triapine are effective regardless of platinum sensitivity status, and promising in combination for both platinum-sensitive and platinum-refractory EOCs.

4.
Br J Cancer ; 114(7): 777-86, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26964031

ABSTRACT

BACKGROUND: Platinum resistance may be attributable to inherent or acquired proficiency in homologous recombination repair (HRR) in epithelial ovarian cancer (EOC). The objective of this study was to evaluate the efficacy of the small molecule inhibitor triapine to disrupt HRR and sensitise BRCA wild-type EOC cells to platinum-based combination therapy in vitro and in vivo. METHODS: The sensitivity of BRCA wild-type cancer cells to olaparib, cisplatin, carboplatin, doxorubicin, or etoposide in combination with triapine was evaluated by clonogenic survival assays. The effects of triapine on HRR activity in cells were measured with a DR-GFP reporter assay. The ability of triapine to enhance the effects of the carboplatin-doxil combination on EOC tumour growth delay was determined using a xenograft tumour mouse model. RESULTS: Platinum resistance is associated with wild-type BRCA status. Triapine inhibits HRR activity and enhances the sensitivity of BRCA wild-type cancer cells to cisplatin, olaparib, and doxorubicin. However, sequential combination of triapine and cisplatin is necessary to achieve synergism. Moreover, triapine potentiates platinum-based combination therapy against BRCA wild-type EOC cells and produces significant delay of EOC tumour growth. CONCLUSIONS: Triapine promises to augment the clinical efficacy of platinum-based combination regimens for treatment of platinum-resistant EOC with wild-type BRCA and proficient HRR activity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Recombinational DNA Repair/drug effects , Animals , Breast Neoplasms/pathology , Carboplatin/administration & dosage , Carcinoma, Ovarian Epithelial , Cisplatin/administration & dosage , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , Female , Humans , Mice , Mice, Nude , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Phthalazines/administration & dosage , Piperazines/administration & dosage , Polyethylene Glycols/administration & dosage , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Biochem Pharmacol ; 91(3): 312-22, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25130544

ABSTRACT

Triapine, currently being evaluated as an antitumor agent in phase II clinical trials, and its terminally dimethylated derivative Dp44mT share the α-pyridyl thiosemicarbazone backbone that functions as ligands for transition metal ions. Yet, Dp44mT is approximately 100-fold more potent than triapine in cytotoxicity assays. The aims of this study were to elucidate the mechanisms underlying their potency disparity and to determine their kinetics of cell-kill in culture to aid in the formulation of their clinical dosing schedules. The addition of Cu(2+) inactivated triapine in a 1:1 stoichiometric fashion, while it potentiated the cytotoxicity of Dp44mT. Clonogenic assays after finite-time drug-exposure revealed that triapine produced cell-kill in two phases, one completed within 20 min that caused limited cell-kill, and the other occurring after 16 h of exposure that produced extensive cell-kill. The ribonucleotide reductase inhibitor triapine at 0.4 µM caused immediate complete arrest of DNA synthesis, whereas Dp44mT at this concentration did not appreciably inhibit DNA synthesis. The inhibition of DNA synthesis by triapine was reversible upon its removal from the medium. Cell death after 16 h exposure to triapine paralleled the appearance of phospho-(γ)H2AX, a marker of DNA double-strand breaks induced by collapse of DNA replication forks after prolonged replication arrest. In contrast to triapine, Dp44mT produced robust cell-kill within 1h in a concentration-dependent manner. The short-term action of both agents was prevented by thiols, indicative of the involvement of reactive oxygen species. The time dependency in the production of cell-kill by triapine should be considered in treatment regimens.


Subject(s)
Antineoplastic Agents/pharmacology , Copper/pharmacology , Pyridines/pharmacology , Thiosemicarbazones/pharmacology , Cell Line, Tumor/drug effects , Cell Survival/drug effects , DNA Breaks, Double-Stranded/drug effects , Enzymes/metabolism , HL-60 Cells/drug effects , Histones/metabolism , Humans , Inhibitory Concentration 50 , Time Factors
6.
Hypertension ; 64(4): 784-91, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24980670

ABSTRACT

The role of salt intake in the development of hypertension is prominent, but its mechanism has not been fully elucidated. Our aim was to examine the effect of transient salt intake during the prehypertensive period in hypertensive model animals. Dahl salt-sensitive rats and spontaneously hypertensive rats were fed from 6 to 14 weeks with low-salt (0.12% NaCl), normal-salt (0.8% NaCl), high-salt (7% NaCl), or high-sodium/normal-chloride diet and returned to normal-salt diet for 3 months. Rats in the high-salt group saw elevations in blood pressure (BP) not only during the treatment period but also for the 3 months after returning to normal-salt diet. We named this phenomenon salt memory. Renal arteriolar injury was found in the high-salt group at the end of experiment. Dahl salt-sensitive rats were fed from 6 to 14 weeks with high-salt diet with angiotensin receptor blocker, vasodilator, calcium channel blocker, and calcium channel blocker+angiotensin receptor blocker and returned to normal-salt diet. Although BP was suppressed to control levels by vasodilator or calcium channel blocker, elevated renal angiotensin II and renal arteriolar injury were observed, and salt memory did not disappear because of sustained renal arteriolar injury. Calcium channel blocker+angiotensin receptor blocker suppressed renal arteriolar injury, resulting in the disappearance of salt memory. Cross-transplantation of kidneys from Dahl salt-sensitive rats on high salt to control rats caused increase of BP, whereas control kidneys caused reduction in BP of hypertensive rats, inducing the central role of the kidney. These results suggest that renal arteriolar injury through BP and renal angiotensin II elevation plays important roles in the development of salt memory for hypertension.


Subject(s)
Arterioles/physiopathology , Blood Pressure/physiology , Hypertension/physiopathology , Kidney/blood supply , Sodium Chloride, Dietary/administration & dosage , Angiotensin II/blood , Angiotensin II/metabolism , Angiotensin Receptor Antagonists/pharmacology , Animals , Arterioles/drug effects , Arterioles/injuries , Blood Pressure/drug effects , Blotting, Western , Calcium Channel Blockers/pharmacology , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Gene Expression/drug effects , Hypertension/etiology , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Kidney Transplantation/methods , Male , Rats , Rats, Inbred Dahl , Rats, Inbred SHR , Renin/blood , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Reverse Transcriptase Polymerase Chain Reaction , Sodium Chloride, Dietary/toxicity , Time Factors
7.
Chem Res Toxicol ; 27(8): 1440-9, 2014 Aug 18.
Article in English | MEDLINE | ID: mdl-25012050

ABSTRACT

Prodrugs of 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine (90CE) are promising anticancer agents. The 90CE moiety is a readily latentiated, short-lived (t1/2 ∼ 30 s) chloroethylating agent that can generate high yields of oxophilic electrophiles responsible for the chloroethylation of the O-6 position of guanine in DNA. These guanine O-6 alkylations are believed to be responsible for the therapeutic effects of 90CE and its prodrugs. Thus, 90CE demonstrates high selectivity toward tumors with diminished levels of O(6)-alkylguanine-DNA alkyltransferase (MGMT), the resistance protein responsible for O(6)-alkylguanine repair. The formation of O(6)-(2-chloroethyl)guanine lesions ultimately leads to the generation of highly cytotoxic 1-(N(3)-cytosinyl),-2-(N(1)-guaninyl)ethane DNA interstrand cross-links via N(1),O(6)-ethanoguanine intermediates. The anticancer activity arising from this sequence of reactions is thus identical to this component of the anticancer activity of the clinically used chloroethylnitrosoureas. Herein, we evaluate the ability of glutathione (GSH) and other low molecular weight thiols, as well as GSH coupled with various glutathione S-transferase enzymes (GSTs) to attenuate the final yields of cross-links generated by 90CE when added prior to or immediately following the initial chloroethylation step to determine the major point(s) of interaction. In contrast to studies utilizing BCNU as a chloroethylating agent by others, GSH (or GSH/GST) did not appreciably quench DNA interstrand cross-link precursors. While thiols alone offered little protection at either alkylation step, the GSH/GST couple was able to diminish the initial yields of cross-link precursors. 90CE exhibited a very different GST isoenzyme susceptibility to that reported for BCNU, this could have important implications in the relative resistance of tumor cells to these agents. The protection afforded by GSH/GST was compared to that produced by MGMT.


Subject(s)
Antineoplastic Agents/chemistry , DNA/chemistry , Glutathione Transferase/metabolism , Glutathione/chemistry , Hydrazines/chemistry , Sulfonamides/chemistry , Alkylation , Animals , Antineoplastic Agents/metabolism , Cell Line, Tumor , DNA/metabolism , DNA Modification Methylases/chemistry , DNA Modification Methylases/metabolism , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/metabolism , Glutathione/metabolism , Glutathione Transferase/genetics , Guanine/chemistry , Guanine/metabolism , Hydrazines/metabolism , Mice , Prodrugs/chemistry , Prodrugs/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Sulfonamides/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism
8.
Chem Res Toxicol ; 27(5): 818-33, 2014 May 19.
Article in English | MEDLINE | ID: mdl-24618018

ABSTRACT

Prodrugs of the short-lived chloroethylating agent 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine (90CE) and its methylating analogue 1,2-bis(methylsulfonyl)-1-(methyl)hydrazine (KS90) are potentially useful anticancer agents. This class of agents frequently yields higher ratios of therapeutically active oxophilic electrophiles responsible for DNA O(6)-guanine alkylations to other electrophiles with lower therapeutic relevance than the nitrosoureas. This results in improved selectivity toward tumors with diminished levels of O(6)-alkylguanine-DNA alkyltransferase (MGMT), the resistance protein responsible for O(6)-alkylguanine repair. The formation of O(6)-(2-chloroethyl)guanine, which leads to the formation of a DNA-DNA interstrand cross-link, accounts for the bulk of the anticancer activity of 90CE prodrugs. Herein, we describe a new decomposition pathway that is available to 90CE but not to its methylating counterpart. This pathway appears to be subject to general/acid base catalysis with phosphate (Pi), phosphomonoesters, and phosphodiesters, being particularly effective. This pathway does not yield a chloroethylating species and results in a major change in nucleophile preference since thiophilic rather than oxophilic electrophiles are produced. Thus, a Pi concentration dependent decrease in DNA-DNA interstand cross-link formation was observed. Changes in 90CE decomposition products but not alkylation kinetics occurred in the presence of Pi since the prebranch point elimination of the N-1 methanesulfinate moiety remained the rate-limiting step. The Pi catalyzed route is expected to dominate at Pi and phosphoester concentrations totaling >25-35 mM. In view of the abundance of Pi and phosphoesters in cells, this pathway may have important effects on agent toxicity, tumor selectivity, and resistance to prodrugs of 90CE. Furthermore, it may be possible to design analogues that diminish this thiophile-generating pathway, which is likely superfluous at best and potentially detrimental to the targeting of hypoxic regions where Pi concentrations can be significantly elevated.


Subject(s)
Antineoplastic Agents/metabolism , Hydrazines/metabolism , Intercalating Agents/metabolism , Prodrugs/metabolism , Sulfonamides/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA/chemistry , DNA/metabolism , Hydrazines/chemistry , Hydrazines/pharmacology , Intercalating Agents/chemistry , Intercalating Agents/pharmacology , Mice , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphates/metabolism , Prodrugs/chemistry , Prodrugs/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology
9.
Mol Carcinog ; 53(3): 201-10, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23065697

ABSTRACT

MGMT is the primary vehicle for cellular removal of alkyl lesions from the O-6 position of guanine and the O-4 position of thymine. While key to the maintenance of genomic integrity, MGMT also removes damage induced by alkylating chemotherapies, inhibiting the efficacy of cancer treatment. Germline variants of human MGMT are well-characterized, but somatic variants found in tumors were, prior to this work, uncharacterized. We found that MGMT G132R, from a human esophageal tumor, and MGMT G156C, from a human colorectal cancer cell line, are unable to rescue methyltransferase-deficient Escherichia coli as well as wild type (WT) human MGMT after treatment with a methylating agent. Using pre-steady state kinetics, we biochemically characterized these variants as having a reduced rate constant. G132R binds DNA containing an O6 -methylguanine lesion half as tightly as WT MGMT, while G156C has a 40-fold decrease in binding affinity for the same damaged DNA versus WT. Mammalian cells expressing either G132R or G156C are more sensitive to methylating agents than mammalian cells expressing WT MGMT. G132R is slightly resistant to O6 -benzylguanine, an inhibitor of MGMT in clinical trials, while G156C is almost completely resistant to this inhibitor. The impared functionality of expressed variants G132R and G156C suggests that the presence of somatic variants of MGMT in a tumor could impact chemotherapeutic outcomes.


Subject(s)
DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , DNA Repair/genetics , Drug Resistance, Neoplasm/genetics , Mammary Neoplasms, Experimental/genetics , Mutation/genetics , Tumor Suppressor Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , DNA Modification Methylases/antagonists & inhibitors , DNA Repair/drug effects , DNA Repair Enzymes/antagonists & inhibitors , Female , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Mammary Neoplasms, Experimental/pathology , Mice , Tumor Cells, Cultured , Tumor Suppressor Proteins/antagonists & inhibitors
10.
J Cancer Ther ; 4(4): 919-931, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23946891

ABSTRACT

The tumor selectivity of alkylating agents that produce guanine O6-chloroethyl (laromustine and carmustine) and O6-methyl (temozolomide) lesions, depends upon O6-methylguanine-DNA methyltransferase (MGMT) activity being lower in tumor than in host tissue. Despite the established role of MGMT as a tumor resistance factor, consensus on how to assess MGMT expression in clinical samples is unsettled. The aim of this study is to examine the relationship between the values derived from distinctive MGMT measurements in 13, 12, 6 and 2 pairs of human tumors and matched normal adjacent tissue from the colon, kidney, lung and liver, respectively, and in human cell lines. The MGMT measurements included (a) alkyl-transfer assays using [benzene-3H]O6-benzylguanine as a substrate to assess functional MGMT activity, (b) methylation-specific PCR (MSP) to probe MGMT gene promoter CpG methylations as a measure of gene silencing, and (c) western immunoblots to analyze the MGMT protein. In human cell lines, a strict negative correlation existed between MGMT activity and the extent of promoter methylation. In tissue specimens, by contrast, the correlation between these two variables was low. Moreover, alkyl-transfer assays identified 3 pairs of tumors and normal tissue with tumor-selective reduction in MGMT activity in the absence of promoter methylation. Cell line MGMT migrated as a single band in western analyses, whereas tissue MGMT was heterogeneous around its molecular size and at much higher molecular masses, indicative of multi-layered post-translational modifications. Malignancy is occasionally associated with a mobility shift in MGMT. Contrary to the prevalent expectation that MGMT expression is governed at the level of gene silencing, these data suggest that other mechanisms that can lead to tumor-selective reduction in MGMT activity exist in human tissue.

11.
Bioorg Med Chem Lett ; 23(6): 1853-9, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23395657

ABSTRACT

Two new agents based upon the structure of the clinically active prodrug laromustine were synthesized. These agents, 2-(2-chloroethyl)-N-methyl-1,2-bis(methylsulfonyl)-N-nitrosohydrazinecarboxamide (1) and N-(2-chloroethyl)-2-methyl-1,2-bis(methylsulfonyl)-N-nitrosohydrazinecarboxamide (2), were designed to retain the potent chloroethylating and DNA cross-linking functions of laromustine, and gain the ability to methylate DNA at the O-6 position of guanine, while lacking the carbamoylating activity of laromustine. The methylating arm was introduced with the intent of depleting the DNA repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT). Compound 1 is markedly more cytotoxic than laromustine in both AGT minus EMT6 mouse mammary carcinoma cells and high AGT expressing DU145 human prostate carcinoma cells. DNA cross-linking studies indicated that its cross-linking efficiency is nearly identical to its predicted active decomposition product, 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine (90CE), which is also produced by laromustine. AGT ablation studies in DU145 cells demonstrated that 1 can efficiently deplete AGT. Studies assaying methanol and 2-chloroethanol production as a consequence of the methylation and chloroethylation of water by 1 and 2 confirmed their ability to function as methylating and chloroethylating agents and provided insights into the superior activity of 1.


Subject(s)
Antineoplastic Agents, Alkylating/chemistry , Methylnitrosourea/analogs & derivatives , Sulfonamides/chemistry , Animals , Antineoplastic Agents, Alkylating/pharmacokinetics , Antineoplastic Agents, Alkylating/toxicity , Cell Line, Tumor , Cell Survival/drug effects , DNA/chemistry , DNA/metabolism , DNA Adducts/chemistry , DNA Methylation , Half-Life , Humans , Hydrazines/chemistry , Hydrazines/pharmacokinetics , Hydrazines/toxicity , Methylnitrosourea/chemical synthesis , Methylnitrosourea/chemistry , Methylnitrosourea/toxicity , Mice , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Sulfonamides/chemical synthesis , Sulfonamides/toxicity
12.
Bioorg Med Chem Lett ; 22(19): 6242-7, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22932317

ABSTRACT

The efficacy of agents that alkylate the O-6 position of guanine is inhibited by O(6)-alkylguanine-DNA alkyltransferase (AGT) which removes these lesions from the tumor DNA. To increase differential toxicity, inhibitors must selectively deplete AGT in tumors, while sparing normal tissues where this protein serves a protective function. A newly synthesized prodrug of the AGT inhibitor O(6)-benzylguanine (O(6)-BG) with an α,α-dimethyl-4-nitrobenzyloxycarbonyl moiety masking the essential 2-amino group has demonstrated the feasibility of targeting hypoxic regions that are unique to solid tumors, for drug delivery. However, these modifications resulted in greatly decreased solubility. Recently, new potent global AGT inhibitors with improved formulatability such as O(6)-[(3-aminomethyl)benzylguanine (1) have been developed. However, acetylamino (N-(3-(((2-amino-9H-purin-6-yl)oxy)methyl)benzyl)acetamide) (2) exhibits a pronounced decrease in activity. Thus, 1 would be inactivated by N-acetylation and probably N-glucuronidation. To combat potential conjugational inactivation while retaining favorable solubility, we synthesized 6-((3-((dimethylamino)methyl)benzyl)oxy)-9H-purin-2-amine (3) in which the 3-aminomethyl moiety is protected by methylation; and to impart tumor selectivity we synthesized 2-(4-nitrophenyl)propan-2-yl(6-((3-((dimethylamino)methyl)benzyl)oxy)-9H-purin-2-yl)carbamate (7), a hypoxia targeted prodrug of 3 utilizing an α,α-dimethyl-4-nitrobenzyloxycarbonyl moiety. Consistent with this design, 7 demonstrates both hypoxia selective conversion by EMT6 cells of 7 to 3 and hypoxic sensitization of AGT containing DU145 cells to the cytotoxic actions of laromustine, while exhibiting improved solubility.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Enzyme Inhibitors/chemical synthesis , Hypoxia , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Prodrugs/chemical synthesis , Prodrugs/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , HL-60 Cells , Humans , Mice , Molecular Structure , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Prodrugs/chemistry , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Solubility , Structure-Activity Relationship
13.
Chem Biol Drug Des ; 80(2): 279-90, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22553921

ABSTRACT

Cellular resistance to chemotherapeutics that alkylate the O-6 position of guanine residues in DNA correlates with their O(6)-alkylguanine-DNA alkyltransferase activity. In normal cells high [O(6)-alkylguanine-DNA alkyltransferase] is beneficial, sparing the host from toxicity, whereas in tumor cells high [O(6)-alkylguanine-DNA alkyltransferase] prevents chemotherapeutic response. Therefore, it is necessary to selectively inactivate O(6)-alkylguanine-DNA alkyltransferase in tumors. The oxygen-deficient compartment unique to solid tumors is conducive to reduction, and could be utilized to provide this selectivity. Therefore, we synthesized 2-nitro-6-benzyloxypurine, an analog of O(6)-benzylguanine in which the essential 2-amino group is replaced by a nitro moiety, and 2-nitro-6-benzyloxypurine is >2000-fold weaker than O(6)-benzylguanine as an O(6)-alkylguanine-DNA alkyltransferase inhibitor. We demonstrate oxygen concentration sensitive net reduction of 2-nitro-6-benzyloxypurine by cytochrome P450 reductase, xanthine oxidase, and EMT6, DU145, and HL-60 cells to yield O(6)-benzylguanine. We show that 2-nitro-6-benzyloxypurine treatment depletes O(6)-alkylguanine-DNA alkyltransferase in intact cells under oxygen-deficient conditions and selectively sensitizes cells to laromustine (an agent that chloroethylates the O-6 position of guanine) under oxygen-deficient but not normoxic conditions. 2-Nitro-6-benzyloxypurine represents a proof of concept lead compound; however, its facile reduction (E(1/2) - 177 mV versus Ag/AgCl) may result in excessive oxidative stress and/or the generation of O(6)-alkylguanine-DNA alkyltransferase inhibitors in normoxic regions in vivo.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Guanine/analogs & derivatives , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Oxygen/metabolism , Antineoplastic Agents/pharmacology , Cell Hypoxia/drug effects , Cell Line , Enzyme Inhibitors/chemical synthesis , Guanine/chemical synthesis , Guanine/chemistry , Guanine/pharmacology , HL-60 Cells , Humans , Hydrazines/pharmacology , Hydrogen Peroxide/metabolism , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Oxidation-Reduction , Purines/chemical synthesis , Purines/chemistry , Purines/pharmacology , Sulfonamides/pharmacology , Superoxides/metabolism
14.
Int J Radiat Biol ; 88(3): 277-85, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22111842

ABSTRACT

PURPOSE: These studies explored questions related to the potential use of Laromustine in the treatment of solid tumors and in combination with radiotherapy. MATERIALS AND METHODS: The studies used mouse EMT6 cells (both parental and transfected with genes for O(6)-alkylguanine-DNA transferase [AGT]), repair-deficient human Fanconi Anemia C and Chinese hamster VC8 (BRCA2(-/-)) cells and corresponding control cells, and EMT6 tumors in mice assayed using cell survival and tumor growth assays. RESULTS: Hypoxia during Laromustine treatment did not protect EMT6 cells or human fibroblasts from this agent. Rapidly proliferating EMT6 cells were more sensitive than quiescent cultures. EMT6 cells expressing mouse or human AGT, which removes O(6)-alkyl groups from DNA guanine, thereby protecting against G-C crosslink formation, increased resistance to Laromustine. Crosslink-repair-deficient Fanconi Anemia C and VC8 cells were hypersensitive to Laromustine, confirming the importance of crosslinks as lethal lesions. In vitro, Laromustine and radiation produced additive toxicities to EMT6 cells. Studies using tumor cell survival and tumor growth assays showed effects of regimens combining Laromustine and radiation that were compatible with additive or subadditive interactions. CONCLUSIONS: The effects of Laromustine on solid tumors and with radiation are complex and are influenced by microenvironmental and proliferative heterogeneity within these malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Hydrazines/pharmacology , Neoplasms/drug therapy , Neoplasms/radiotherapy , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Combined Modality Therapy , Cricetinae , DNA Repair/drug effects , DNA Repair/radiation effects , Humans , Hydrazines/therapeutic use , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/radiotherapy , Mice , Neoplasms/metabolism , Neoplasms/pathology , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Radiation Tolerance/drug effects , Sulfonamides/therapeutic use , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects , Xenograft Model Antitumor Assays
15.
Chem Biol Drug Des ; 78(4): 513-26, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21777394

ABSTRACT

The anticancer prodrug 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)-2-[[1-(4-nitrophenyl)ethoxy]carbonyl]hydrazine (KS119) selectively releases a short-lived cytotoxin following enzymatic reduction in hypoxic environments found in solid tumors. KS119, in addition to two enantiomers, has two stable atropisomers (conformers differing in structure owing to hindered bond rotation) that interconvert at 37 °C in aqueous solution by first-order kinetics with t(1/2) values of ∼50 and ∼64 h. The atropisomers differ in physical properties such as partition coefficients that allow their chromatographic separation on non-chiral columns. A striking difference in the rate of metabolism of the two atropisomers occurs in intact EMT6 murine mammary carcinoma cells under oxygen-deficient conditions. A structurally related molecule, 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)-2-[[1-(3-hydroxy-4-nitrophenyl)ethoxy]carbonyl]hydrazine (KS119WOH), was also found to exist in similar stable atropisomers. The ratio of the atropisomers of KS119 and structurally related agents has the potential to impact the bioavailability, activation, and therapeutic activity. Thus, thermally stable atropisomers/conformers in small molecules can result in chemically and enantiomerically pure compounds having differences in biological activities.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Hydrazines/chemistry , Hydrazines/pharmacology , Mammary Neoplasms, Animal/drug therapy , Prodrugs/chemistry , Prodrugs/pharmacology , Animals , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Female , Hydrazines/metabolism , Isomerism , Mammary Neoplasms, Animal/metabolism , Mice , Models, Molecular , Molecular Conformation , Prodrugs/metabolism
16.
Biochem Pharmacol ; 81(10): 1201-10, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21396917

ABSTRACT

To most effectively treat cancer it may be necessary to preferentially destroy tumor tissue while sparing normal tissues. One strategy to accomplish this is to selectively cripple the involved tumor resistance mechanisms, thereby allowing the affected anticancer drugs to gain therapeutic efficacy. Such an approach is exemplified by our design and synthesis of the intracellular hypoxic cell activated methylating agent, 1,2-bis(methylsulfonyl)-1-methyl-2-[[1-(4-nitrophenyl)ethoxy]carbonyl]hydrazine (KS900) that targets the O-6 position of guanine in DNA. KS900 is markedly more cytotoxic in clonogenic experiments under conditions of oxygen deficiency than the non-intracellularly activated agents KS90, and 90M, when tested in O(6)-alkylguanine-DNA alkyltransferase (AGT) non-expressing cells (EMT6 mouse mammary carcinoma, CHO/AA8 hamster ovary, and U251 human glioma), and than temozolomide when tested in AGT expressing cells (DU145 human prostate carcinoma). Furthermore, KS900 more efficiently ablates AGT in HL-60 human leukemia and DU145 cells than the spontaneous globally activated methylating agent KS90, with an IC(50) value over 9-fold lower than KS90. Finally, KS900 under oxygen-deficient conditions selectively sensitizes DU145 cells to the chloroethylating agent, onrigin, through the ablation of the resistance protein AGT. Thus, under hypoxia, KS900 is more cytotoxic at substantially lower concentrations than methylating agents such as temozolomide that are not preferentially activated in neoplastic cells by intracellular reductase catalysts. The necessity for intracellular activation of KS900 permits substantially greater cytotoxic activity against cells containing the resistance protein O(6)-alkylguanine-DNA alkyltransferase (AGT) than agents such as temozolomide. Furthermore, the hypoxia-directed intracellular activation of KS900 allows it to preferentially ablate AGT pools under the oxygen-deficient conditions that are present in malignant tissue.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Hydrazines/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Prodrugs/pharmacology , Sulfonamides/pharmacology , Animals , Cell Hypoxia , Cell Line, Tumor , Cricetinae , Cricetulus , Humans , Methylation , Mice , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Oxidoreductases/metabolism
17.
Biochem Pharmacol ; 80(9): 1317-25, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20654586

ABSTRACT

O(6)-Alkylguanine-DNA alkyltransferase (AGT) mediates tumor resistance to alkylating agents that generate guanine O(6)-chloroethyl (Onrigin™ and carmustine) and O(6)-methyl (temozolomide) lesions; however, the relative efficiency of AGT protection against these lesions and the degree of resistance to these agents that a given number of AGT molecules produces are unclear. Measured from differential cytotoxicity in AGT-ablated and AGT-intact HL-60 cells containing 17,000 AGT molecules/cell, AGT produced 12- and 24-fold resistance to chloroethylating (90CE) and methylating (KS90) analogs of Onrigin™, respectively. For 50% growth inhibition, KS90 and 90CE generated 5,600 O(6)-methylguanines/cell and ∼300 O(6)-chloroethylguanines/cell, respectively. AGT repaired O(6)-methylguanines until the AGT pool was exhausted, while its repair of O(6)-chloroethylguanines was incomplete due to progression of the lesions to AGT-irreparable interstrand DNA cross-links. Thus, the smaller number of O(6)-chloroethylguanine lesions needed for cytotoxicity accounted for the marked degree of resistance (12-fold) to 90CE produced by AGT. Transfection of human or murine AGT into AGT deficient transplantable tumor cells (i.e., EMT6, M109 and U251) generated transfectants expressing AGT ranging from 4,000 to 700,000 molecules/cell. In vitro growth inhibition assays using these transfectants treated with 90CE revealed that AGT caused a concentration dependent resistance up to a level of ∼10,000 AGT molecules/cell. This finding was corroborated by in vivo studies where expression of 4,000 and 10,000 murine AGT molecules/cell rendered EMT6 tumors partially and completely resistant to Onrigin™, respectively. These studies imply that the antitumor activity of Onrigin™ stems from guanine O(6)-chloroethylation and define the threshold concentration of AGT that negates its antineoplastic activity.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage , Guanine/analogs & derivatives , Hydrazines/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/physiology , Sulfonamides/pharmacology , Animals , Carmustine/pharmacology , Cell Line, Tumor , DNA Methylation , DNA Repair , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Drug Resistance, Neoplasm , Guanine/analysis , Humans , Mice , O(6)-Methylguanine-DNA Methyltransferase/analysis , Temozolomide
18.
Nephron Exp Nephrol ; 115(2): e22-32, 2010.
Article in English | MEDLINE | ID: mdl-20407279

ABSTRACT

BACKGROUND/AIMS: The role of matrix metalloproteinases (MMPs) in the pathogenesis of glomerular injury appears to be complex. To investigate the role of individual MMPs, we examined the course of Adriamycin-induced albuminuria and glomerulosclerosis in mice lacking either a gelatinase (MMP-9) or a collagenase (MMP-13). METHODS: Adriamycin was administered to MMP-9 or MMP-13 knockout (KO) mice. Glomerular injury was assessed by the quantification of albuminuria, the glomerular injury score and type IV collagen immunostaining. RESULTS: Treatment of mice with Adriamycin (18 mg/kg i.v.) resulted in marked albuminuria and glomerulosclerosis reaching a peak at 4-8 weeks. The albuminuria and glomerulosclerosis were significantly (p < 0.05) attenuated in both the MMP-9 KO and MMP-13 KO mice compared to controls. In contrast, treatment of wild-type mice with the broad-spectrum MMP inhibitor doxycycline did not have a beneficial effect on the albuminuria and glomerulosclerosis. CONCLUSION: These results support a role for both gelatinase (MMP-9) and collagenase (MMP-13) in the pathogenesis of glomerular injury in the Adriamycin-induced glomerulosclerosis model. MMP inhibitors with high specificity towards MMP-9 and/or MMP-13 may be potential future candidates to provide more effective therapies to inhibit the development of glomerulosclerosis.


Subject(s)
Albuminuria/chemically induced , Doxorubicin/toxicity , Glomerulosclerosis, Focal Segmental/chemically induced , Matrix Metalloproteinase 13/physiology , Matrix Metalloproteinase 9/physiology , Albuminuria/drug therapy , Albuminuria/enzymology , Albuminuria/prevention & control , Animals , Computer Systems , Doxycycline/pharmacology , Doxycycline/therapeutic use , Drug Evaluation, Preclinical , Enzyme Induction/drug effects , Glomerulosclerosis, Focal Segmental/drug therapy , Glomerulosclerosis, Focal Segmental/enzymology , Glomerulosclerosis, Focal Segmental/prevention & control , Kidney Glomerulus/enzymology , Male , Matrix Metalloproteinase 13/deficiency , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 9/deficiency , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , Random Allocation , Reverse Transcriptase Polymerase Chain Reaction
19.
Kidney Int ; 78(1): 69-78, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20375993

ABSTRACT

Understanding mechanisms that contribute to the regression of glomerulosclerosis is important for developing new strategies to treat chronic kidney disease. We reported that transient high-dose treatment with an angiotensin receptor blocker causes regression of renal arteriolar hypertrophy and hypertension in spontaneously hypertensive rats. To extend those findings to another form of kidney disease, we examined the short- and long-term effects of transient high-dose angiotensin receptor blocker treatment in a mouse model of adriamycin-induced glomerulosclerosis. A 2-week course of candesartan caused a dose-dependent regression of established glomerulosclerotic lesions sustained for over 6 months following cessation of treatment. Highly sensitive in situ zymography and activity assays showed that glomerular matrix metalloproteinase (MMP)-2 activity was increased after high-dose angiotensin blocker therapy. Treatment of cultured podocytes with candesartan resulted in an increase in MMP-2 activity. The regression of glomerulosclerosis was partially attenuated in mice pretreated with the MMP inhibitor doxycycline, as well as in MMP-2 knockout mice. Our results suggest that transient high-dose angiotensin receptor blocker treatment effectively induced sustained regression of glomerulosclerosis by a mechanism mediated, in part, by changes in MMP-2 activity.


Subject(s)
Angiotensin Receptor Antagonists , Kidney Diseases/drug therapy , Kidney Diseases/physiopathology , Matrix Metalloproteinase Inhibitors , Angiotensin II/pharmacology , Angiotensin II/therapeutic use , Angiotensins/pharmacology , Angiotensins/therapeutic use , Animals , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Benzimidazoles , Biphenyl Compounds , Hypertension/drug therapy , Hypertension/pathology , Hypertension/physiopathology , Kidney/drug effects , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/pathology , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Matrix Metalloproteinase 2/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Random Allocation , Rats , Rats, Inbred SHR , Receptors, Angiotensin/therapeutic use , Tetrazoles , Time Factors
20.
Biochem Pharmacol ; 79(11): 1553-61, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20005211

ABSTRACT

1,2-Bis(methylsulfonyl)-1-(2-chloroethyl)-2-[[1-(4-nitrophenyl)ethoxy]carbonyl]hydrazine (KS119) is a prodrug of the 1,2-bis(sulfonyl)hydrazine class of antineoplastic agents designed to exploit the oxygen-deficient regions of cancerous tissue. Thus, under reductive conditions in hypoxic cells this agent decomposes to produce the reactive intermediate 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine (90CE), which in turn generates products that alkylate the O(6)-position of guanine in DNA. Comparison of the cytotoxicity of KS119 in cultured cells lacking O(6)-alkylguanine-DNA alkyltransferase (AGT) to an agent such as Onrigin, which through base catalyzed activation produces the same critical DNA G-C cross-link lesions by the generation of 90CE, indicates that KS119 is substantially more potent than Onrigin under conditions of oxygen deficiency, despite being incompletely activated. In cell lines expressing relatively large amounts of AGT, the design of the prodrug KS119, which requires intracellular activation by reductase enzymes to produce a cytotoxic effect, results in an ability to overcome resistance derived from the expression of AGT. This appears to derive from the ability of a small portion of the chloroethylating species produced by the activation of KS119 to slip through the cellular protection afforded by AGT to generate the few DNA G-C cross-links that are required for tumor cell lethality. The findings also demonstrate that activation of KS119 under oxygen-deficient conditions is ubiquitous, occurring in all of the cell lines tested thus far, suggesting that the enzymes required for reductive activation of this agent are widely distributed in many different tumor types.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Hydrazines/pharmacology , Hypoxia , O(6)-Methylguanine-DNA Methyltransferase/physiology , Prodrugs/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Humans , Hydrazines/pharmacokinetics , O(6)-Methylguanine-DNA Methyltransferase/deficiency , Oxidation-Reduction , Oxidoreductases/metabolism , Prodrugs/metabolism , Prodrugs/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...