Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 130(1): 258-271, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31581151

ABSTRACT

Potentiating radiotherapy and chemotherapy by inhibiting DNA damage repair is proposed as a therapeutic strategy to improve outcomes for patients with solid tumors. However, this approach risks enhancing normal tissue toxicity as much as tumor toxicity, thereby limiting its translational impact. Using NU5455, a newly identified highly selective oral inhibitor of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) activity, we found that it was indeed possible to preferentially augment the effect of targeted radiotherapy on human orthotopic lung tumors without influencing acute DNA damage or a late radiation-induced toxicity (fibrosis) to normal mouse lung. Furthermore, while NU5455 administration increased both the efficacy and the toxicity of a parenterally administered topoisomerase inhibitor, it enhanced the activity of doxorubicin released locally in liver tumor xenografts without inducing any adverse effect. This strategy is particularly relevant to hepatocellular cancer, which is treated clinically with localized drug-eluting beads and for which DNA-PKcs activity is reported to confer resistance to treatment. We conclude that transient pharmacological inhibition of DNA-PKcs activity is effective and tolerable when combined with localized DNA-damaging therapies and thus has promising clinical potential.


Subject(s)
Carcinoma, Hepatocellular , DNA-Activated Protein Kinase/antagonists & inhibitors , Liver Neoplasms, Experimental , Neoplasm Proteins/antagonists & inhibitors , Protein Kinase Inhibitors , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , DNA-Activated Protein Kinase/metabolism , Doxorubicin/pharmacology , Humans , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/pathology , MCF-7 Cells , Mice , Neoplasm Proteins/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays
2.
NMR Biomed ; 30(9)2017 Sep.
Article in English | MEDLINE | ID: mdl-28543772

ABSTRACT

Poly(adenosine diphosphate ribose) polymerases (PARPs) are multifunctional proteins which play a role in many cellular processes. Namely, PARP1 and PARP2 have been shown to be involved in DNA repair, and therefore are valid targets in cancer treatment with PARP inhibitors, such as rucaparib, currently in clinical trials. Proton magnetic resonance spectroscopy (1 H-MRS) was used to study the impact of rucaparib in vitro and ex vivo in liver tissue from mice, via quantitative analysis of nicotinamide adenosine diphosphate (NAD+ ) spectra, to assess the potential of MRS as a biomarker of the PARP inhibitor response. SW620 (colorectal) and A2780 (ovarian) cancer cell lines, and PARP1 wild-type (WT) and PARP1 knock-out (KO) mice, were treated with rucaparib, temozolomide (methylating agent) or a combination of both drugs. 1 H-MRS spectra were obtained from perchloric acid extracts of tumour cells and mouse liver. Both cell lines showed an increase in NAD+ levels following PARP inhibitor treatment in comparison with temozolomide treatment. Liver extracts from PARP1 WT mice showed a significant increase in NAD+ levels after rucaparib treatment compared with untreated mouse liver, and a significant decrease in NAD+ levels in the temozolomide-treated group. The combination of rucaparib and temozolomide did not prevent the NAD+ depletion caused by temozolomide treatment. The 1 H-MRS results show that NAD+ levels can be used as a biomarker of PARP inhibitor and methylating agent treatments, and suggest that in vivo measurement of NAD+ would be valuable.


Subject(s)
Indoles/pharmacology , Liver/metabolism , NAD/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Proton Magnetic Resonance Spectroscopy/methods , Animals , Cell Line, Tumor , Humans , Mice, Knockout , NAD/chemistry
3.
Mol Cancer Ther ; 12(6): 959-67, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23512991

ABSTRACT

Ataxia telangiectasia mutated (ATM) kinase signals DNA double-strand breaks (DSB) to cell-cycle arrest via p53 and DNA repair. ATM-defective cells are sensitive to DSB-inducing agents, making ATM an attractive target for anticancer chemo- and radiosensitization. KU59403 is an ATM inhibitor with the potency, selectivity, and solubility for advanced preclinical evaluation. KU59403 was not cytotoxic to human cancer cell lines (SW620, LoVo, HCT116, and MDA-MB-231) per se but significantly increased the cytotoxicity of topoisomerase I and II poisons: camptothecin, etoposide, and doxorubicin. Chemo- and radiosensitization by ATM inhibition was not p53-dependent. Following administration to mice, KU59403 distributed to tissues and concentrations exceeding those required for in vitro activity were maintained for at least 4 hours in tumor xenografts. KU59403 significantly enhanced the antitumor activity of topoisomerase poisons in mice bearing human colon cancer xenografts (SW620 and HCT116) at doses that were nontoxic alone and well-tolerated in combination. Chemosensitization was both dose- and schedule-dependent. KU59403 represents a major advance in ATM inhibitor development, being the first compound to show good tissue distribution and significant chemosensitization in in vivo models of human cancer, without major toxicity. KU59403 provides the first proof-of-principle preclinical data to support the future clinical development of ATM inhibitors.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Heterocyclic Compounds, 3-Ring/administration & dosage , Neoplasms/drug therapy , Pyrones/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Tumor Suppressor Protein p53/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Camptothecin/administration & dosage , Clinical Trials as Topic , DNA-Binding Proteins , Doxorubicin/administration & dosage , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/drug effects , Etoposide/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Mice , Neoplasms/metabolism , Neoplasms/pathology , Xenograft Model Antitumor Assays
4.
Mol Cancer Ther ; 10(12): 2320-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21926192

ABSTRACT

AG014699 was the first inhibitor of the DNA repair enzyme PARP-1 to enter clinical trial in cancer patients. In addition to enhancing the cytotoxic effect of DNA-damaging chemotherapies, we have previously shown that AG014699 is vasoactive, thereby having the potential to improve drug biodistribution. The effectiveness of the clinical agent doxorubicin is confounded both by poor tumor penetration and cardiotoxicity elicited via PARP hyperactivation. In this study, we analyzed the impact of AG014699 on doxorubicin tolerance and response in breast (MDA-MB-231) and colorectal (SW620, LoVo) tumor models in vitro and in vivo. As anticipated, AG014699 did not potentiate the response to doxorubicin in vitro. In vivo, AG014699 did not influence the pharmacokinetics of doxorubicin; however, it did ameliorate cardiotoxicity. Both toxicity and extent of amelioration were more pronounced in male than in female mice. AG014699 improved vessel perfusion in both MDA-MB-231 and SW620 tumors; however, this neither led to improved tumor-accumulation of doxorubicin nor enhanced therapeutic response. In contrast, when combined with radiotherapy, AG014699 significantly enhanced response both in vitro and in vivo. Real-time assessment of tumor vessel function and companion histologic studies indicate that doxorubicin causes a profound antivascular effect that counters the positive effect of AG014699 on perfusion. These data indicate that although AG014699 can enhance response to some chemotherapeutic drugs via improved delivery, this does not apply to doxorubicin. PARP inhibitors may still be of use to counter doxorubicin toxicity, and if the gender effect translates from rodents to humans, this would have greater effect in males.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Indoles/administration & dosage , Neoplasms/drug therapy , Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cardiotoxins/antagonists & inhibitors , Cell Line, Tumor , Cytoprotection/drug effects , Doxorubicin/antagonists & inhibitors , Drug Synergism , Female , Heart/drug effects , Humans , Indoles/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Neoplasms/blood supply , Neoplasms/metabolism , Perfusion , Poly(ADP-ribose) Polymerase Inhibitors , Treatment Outcome , Xenograft Model Antitumor Assays
5.
J Natl Cancer Inst ; 103(4): 334-46, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21183737

ABSTRACT

BACKGROUND: Mutations in BRCA1 and BRCA2 (BRCA1/2), components of the homologous recombination DNA repair (HRR) pathway, are associated with hereditary breast and ovarian cancers. Poly(ADP-ribose) polymerase (PARP) inhibitors are selectively cytotoxic to animal cells with defective HRR, but results in human cancer cells have been contradictory. We undertook, to our knowledge, the first comprehensive in vitro and in vivo investigations of the antitumor activity of the PARP inhibitor AG014699 in human cancer cells carrying mutated or epigenetically silenced BRCA1/2. METHODS: We used nine human cell lines, four with nonmutated BRCA1/2 (MCF7, MDA-MB-231, and HCC1937-BRCA1 [breast cancer] and OSEC-2 [ovarian surface epithelial]), two with mutated BRCA1 (MDA-MB-436 and HCC1937 [breast cancer]), one with mutated BRCA2 (CAPAN-1 [pancreatic cancer]), one that was heterozygous for BRCA2 (OSEC-1 [ovarian surface epithelial]), and one with epigenetically silenced BRCA1 (UACC3199 [breast cancer]), and two Chinese hamster ovary cell lines, parental AA8 and XRCC3 mutated IRS 1SF. We assessed cytotoxicity, DNA damage, and HRR function. Antitumor activity of AG014699 was determined by growth of xenograft tumors (five mice per treatment group). Long-term safety of AG014699 was assessed. RESULTS: AG014699 (≤10 µM) was cytotoxic to cells with mutated BRCA1/2 or XRCC3 and to UACC3199 cells with epigenetically silenced BRCA1 but not to cells without BRCA1/2 or XRCC3 mutations or that were heterozygous for BRCA2 mutation. AG014699 induced DNA double-strand breaks in all nine cell lines studied. HRR was observed only in cells with functional BRCA1/2 proteins. Growth of xenograft tumors with BRCA1/2 mutations or with epigenetically silenced BRCA1 was reduced by AG014699 treatment, and combination treatment with AG014699 plus carboplatin was more effective than either drug alone. AG014699 was not toxic in mice with nonmutated or heterozygous BRCA2. CONCLUSION: Human cancer cells or xenograft tumors with mutated or epigenetically silenced BRCA1/2 were sensitive to AG014699 monotherapy, indicating a potential role for PARP inhibitors in sporadic human cancers.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Methylation , Genes, BRCA1 , Genes, BRCA2 , Indoles/pharmacology , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , CHO Cells , Carboplatin/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cricetinae , Cricetulus , Enzyme Inhibitors/pharmacology , Female , Gene Silencing , Humans , Mice , Mice, Nude , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Poly(ADP-ribose) Polymerases/metabolism , Transplantation, Heterologous
6.
Cancer Res ; 70(15): 6268-76, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20631063

ABSTRACT

Familial breast and ovarian cancers are often defective in homologous recombination (HR) due to mutations in the BRCA1 or BRCA2 genes. Cisplatin chemotherapy or poly(ADP-ribose) polymerase (PARP) inhibitors were tested for these tumors in clinical trials. In a screen for novel drugs that selectively kill BRCA2-defective cells, we identified 6-thioguanine (6TG), which induces DNA double-strand breaks (DSB) that are repaired by HR. Furthermore, we show that 6TG is as efficient as a PARP inhibitor in selectively killing BRCA2-defective tumors in a xenograft model. Spontaneous BRCA1-defective mammary tumors gain resistance to PARP inhibitors through increased P-glycoprotein expression. Here, we show that 6TG efficiently kills such BRCA1-defective PARP inhibitor-resistant tumors. We also show that 6TG could kill cells and tumors that have gained resistance to PARP inhibitors or cisplatin through genetic reversion of the BRCA2 gene. Although HR is reactivated in PARP inhibitor-resistant BRCA2-defective cells, it is not fully restored for the repair of 6TG-induced lesions. This is likely to be due to several recombinogenic lesions being formed after 6TG. We show that BRCA2 is also required for survival from mismatch repair-independent lesions formed by 6TG, which do not include DSBs. This suggests that HR is involved in the repair of 6TG-induced DSBs as well as mismatch repair-independent 6TG-induced DNA lesion. Altogether, our data show that 6TG efficiently kills BRCA2-defective tumors and suggest that 6TG may be effective in the treatment of advanced tumors that have developed resistance to PARP inhibitors or platinum-based chemotherapy.


Subject(s)
BRCA2 Protein/deficiency , Colonic Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Thioguanine/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis Regulatory Proteins , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Base Pair Mismatch , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , DNA Repair , Drug Synergism , Genes, BRCA2 , HCT116 Cells , Humans , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/genetics , Mice
7.
Clin Cancer Res ; 16(8): 2344-51, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20371688

ABSTRACT

PURPOSE: Poly(ADP-ribose) polymerase (PARP) inhibitors selectively target homologous recombination (HR)-defective cells and show good clinical activity in hereditary breast and ovarian cancer associated with BRCA1 or BRCA2 mutations. A high proportion (up to 50%) of sporadic epithelial ovarian cancers (EOC) could be deficient in HR due to genetic or epigenetic inactivation of BRCA1/BRCA2 or other HR genes. Therefore, there is a potential for extending the use of PARP inhibitors to these patients if HR status can be identified. We developed a functional assay of HR status in primary cultures of EOCs based on Rad51 focus formation that correlates well with sensitivity to the potent PARP inhibitor AG014699. EXPERIMENTAL DESIGN: Primary cultures were derived from ascitic fluid from patients with EOCs. HR status was investigated by gammaH2AX and Rad51 focus formation by immunofluorescence. Cytotoxicity to PARP inhibitors was tested by sulforhodamine B and survival assay. RESULTS: Twenty-five cultures were evaluated for HR status and cytotoxicity to PARP inhibitor. Following exposure to AG014699, there was an increase in Rad51 foci (HR competent) in 9 of 24 (36%) but no increase (HR deficient) in 16 of 24 (64%) cultures. Cytotoxicity was observed in 15 of 16 (93%) HR-deficient samples but not in 9 of 9 HR-competent samples following 24-hour exposure to 10 mumol/L AG014699. CONCLUSION: HR status can be determined in primary cancer samples by Rad51 focus formation, and this correlates with in vitro response to PARP inhibition. Use of this assay as a biomarker now needs testing in the setting of a clinical trial.


Subject(s)
Indoles/pharmacology , Ovarian Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors , Recombination, Genetic , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Biological Assay , Biomarkers, Tumor/metabolism , Cell Survival , Drug Resistance, Neoplasm , Female , Histones/metabolism , Humans , Ovarian Neoplasms/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Rad51 Recombinase/metabolism , Rhodamines/metabolism , Tumor Cells, Cultured
8.
Clin Cancer Res ; 15(19): 6106-12, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19789326

ABSTRACT

PURPOSE: Poly(ADP-ribose) polymerase (PARP) plays an important role in DNA repair, and PARP inhibitors can enhance the activity of DNA-damaging agents in vitro and in vivo. AG014699 is a potent PARP inhibitor in phase II clinical development. However, the range of therapeutics with which AG014699 could interact via a DNA-repair based mechanism is limited. We aimed to investigate a novel, vascular-based activity of AG014699, underlying in vivo chemosensitization, which could widen its clinical application. EXPERIMENTAL DESIGN: Temozolomide response was analyzed in vitro and in vivo. Vessel dynamics were monitored using "mismatch" following the administration of perfusion markers and real-time analysis of fluorescently labeled albumin uptake in to tumors established in dorsal window chambers. Further mechanistic investigations used ex vivo assays of vascular smooth muscle relaxation, gut motility, and myosin light chain kinase (MLCK) inhibition. RESULTS: AG014699 failed to sensitize SW620 cells to temozolomide in vitro but induced pronounced enhancement in vivo. AG014699 (1 mg/kg) improved tumor perfusion comparably with the control agents nicotinamide (1 g/kg) and AG14361 (forerunner to AG014699; 10 mg/kg). AG014699 and AG14361 relaxed preconstricted vascular smooth muscle more potently than the standard agent, hydralazine, with no impact on gut motility. AG014699 inhibited MLCK at concentrations that relaxed isolated arteries, whereas AG14361 had no effect. CONCLUSION: Increased vessel perfusion elicited by AG014699 could increase tumor drug accumulation and therapeutic response. Vasoactive concentrations of AG014699 do not cause detrimental side effects to gut motility and may increase the range of therapeutics with which AG014699 could be combined with for clinical benefit.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blood Vessels/drug effects , Drug Resistance, Neoplasm/drug effects , Indoles/pharmacology , Neoplasms/blood supply , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Azulenes/administration & dosage , Azulenes/pharmacology , Benzodiazepines/administration & dosage , Benzodiazepines/pharmacology , Blood Vessels/physiopathology , Capillary Permeability/drug effects , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Female , HT29 Cells , Humans , Indoles/administration & dosage , Mice , Mice, Nude , Neoplasms/pathology , Temozolomide , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Mol Cancer Ther ; 6(3): 945-56, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17363489

ABSTRACT

Poly(ADP-ribose) polymerase (PARP)-1 (EC 2.4.2.30) is a nuclear enzyme that promotes the base excision repair of DNA breaks. Inhibition of PARP-1 enhances the efficacy of DNA alkylating agents, topoisomerase I poisons, and ionizing radiation. Our aim was to identify a PARP inhibitor for clinical trial from a panel of 42 potent PARP inhibitors (K(i), 1.4-15.1 nmol/L) based on the quinazolinone, benzimidazole, tricyclic benzimidazole, tricyclic indole, and tricyclic indole-1-one core structures. We evaluated chemosensitization of temozolomide and topotecan using LoVo and SW620 human colorectal cells; in vitro radiosensitization was measured using LoVo cells, and the enhancement of antitumor activity of temozolomide was evaluated in mice bearing SW620 xenografts. Excellent chemopotentiation and radiopotentiation were observed in vitro, with 17 of the compounds causing a greater temozolomide and topotecan sensitization than the benchmark inhibitor AG14361 and 10 compounds were more potent radiosensitizers than AG14361. In tumor-bearing mice, none of the compounds were toxic when given alone, and the antitumor activity of the PARP inhibitor-temozolomide combinations was unrelated to toxicity. Compounds that were more potent chemosensitizers in vivo than AG14361 were also more potent in vitro, validating in vitro assays as a prescreen. These studies have identified a compound, AG14447, as a PARP inhibitor with outstanding in vivo chemosensitization potency at tolerable doses, which is at least 10 times more potent than the initial lead, AG14361. The phosphate salt of AG14447 (AG014699), which has improved aqueous solubility, has been selected for clinical trial.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Cell Line, Tumor/drug effects , Cell Line, Tumor/radiation effects , Colorectal Neoplasms/radiotherapy , DNA Repair/drug effects , DNA Repair/radiation effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Gamma Rays , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Heterocyclic Compounds, 3-Ring/chemistry , Humans , Maximum Tolerated Dose , Mice , Mice, Nude , Poly(ADP-ribose) Polymerases/metabolism , Structure-Activity Relationship , Temozolomide , Topoisomerase I Inhibitors , Topotecan/pharmacology
10.
Nature ; 434(7035): 913-7, 2005 Apr 14.
Article in English | MEDLINE | ID: mdl-15829966

ABSTRACT

Poly(ADP-ribose) polymerase (PARP1) facilitates DNA repair by binding to DNA breaks and attracting DNA repair proteins to the site of damage. Nevertheless, PARP1-/- mice are viable, fertile and do not develop early onset tumours. Here, we show that PARP inhibitors trigger gamma-H2AX and RAD51 foci formation. We propose that, in the absence of PARP1, spontaneous single-strand breaks collapse replication forks and trigger homologous recombination for repair. Furthermore, we show that BRCA2-deficient cells, as a result of their deficiency in homologous recombination, are acutely sensitive to PARP inhibitors, presumably because resultant collapsed replication forks are no longer repaired. Thus, PARP1 activity is essential in homologous recombination-deficient BRCA2 mutant cells. We exploit this requirement in order to kill BRCA2-deficient tumours by PARP inhibition alone. Treatment with PARP inhibitors is likely to be highly tumour specific, because only the tumours (which are BRCA2-/-) in BRCA2+/- patients are defective in homologous recombination. The use of an inhibitor of a DNA repair enzyme alone to selectively kill a tumour, in the absence of an exogenous DNA-damaging agent, represents a new concept in cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , BRCA2 Protein/deficiency , Genes, BRCA2 , Neoplasms/drug therapy , Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Azulenes , BRCA2 Protein/genetics , Benzodiazepines/pharmacology , Cell Line, Tumor , DNA Damage , DNA Repair , DNA Replication , Mice , Mice, Nude , Neoplasms/enzymology , Neoplasms/pathology , Poly(ADP-ribose) Polymerases/deficiency , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Xenograft Model Antitumor Assays
11.
J Med Chem ; 47(22): 5467-81, 2004 Oct 21.
Article in English | MEDLINE | ID: mdl-15481984

ABSTRACT

The design, synthesis, and biological evaluation of potent inhibitors of poly(ADP-ribose) polymerase-1 (PARP-1) are reported. A novel series of 3,4-dihydro-2H-[1,4]diazepino[6,7,1-hi]indol-1-ones were designed using a combination of protein structure-based drug design, molecular modeling, and structure-activity relationships (SAR). These novel submicromolar inhibitors possess a tricyclic ring system conformationally restricting the benzamide in the preferred cis orientation. The compounds were designed to optimize space-filling and atomic interactions within the NAD+ binding site of PARP-1. Previously described and newly adapted methods were applied to syntheses of these tricyclic inhibitors. Various modifications were made to the diazepinoindolones at the 6- and 7-positions in order to study this region of the active site and optimize noncovalent interactions. The electron density of derivative 28 bound to chicken PARP-1 revealed that the oxime makes a tight hydrogen bond with the catalytic gamma-carboxylate of glutamic acid (Glu) 988 in accordance with our original designs and models. Most of the compounds have been evaluated for inhibition of human PARP-1. Selected inhibitors were also tested for the ability to potentiate the cytotoxic effect of the DNA-damaging agent Topotecan.


Subject(s)
Antineoplastic Agents/chemical synthesis , Azepines/chemical synthesis , Indoles/chemical synthesis , Poly(ADP-ribose) Polymerase Inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Azepines/chemistry , Azepines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Design , Drug Resistance, Neoplasm , Drug Synergism , Humans , Indoles/chemistry , Indoles/pharmacology , Models, Molecular , Structure-Activity Relationship , Topoisomerase I Inhibitors
12.
Bioorg Med Chem Lett ; 14(10): 2433-7, 2004 May 17.
Article in English | MEDLINE | ID: mdl-15109627

ABSTRACT

The synthesis and biological evaluation of a new series of amine-substituted 2-arylbenzimidazole-4-carboxamide inhibitors of the DNA-repair enzyme poly(ADP-ribose) polymerase-1 (PARP-1) is reported. The introduction of an amine substituent at the 2-aryl position is not detrimental to activity, with most inhibitors exhibiting K(i) values for PARP-1 inhibition in the low nanomolar range. Two compounds in this series were found to potentiate the cytotoxicity of the DNA-methylating agent temozolomide by 4-5-fold in a human colorectal cancer cell line.


Subject(s)
Amides/pharmacology , Antineoplastic Agents/chemical synthesis , Benzimidazoles/pharmacology , Dacarbazine/analogs & derivatives , Poly(ADP-ribose) Polymerase Inhibitors , Amides/chemical synthesis , Animals , Antineoplastic Agents/pharmacology , Benzimidazoles/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Dacarbazine/pharmacology , Drug Synergism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Structure-Activity Relationship , Temozolomide
13.
Clin Cancer Res ; 10(3): 881-9, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871963

ABSTRACT

PURPOSE: Mismatch repair (MMR) deficiency confers resistance to temozolomide, a clinically active DNA-methylating agent. The purpose of the current study was to investigate the reversal mechanism of temozolomide resistance by the potent novel poly(ADP-ribose) polymerase (PARP)-1 inhibitor, AG14361, in MMR-proficient and -deficient cells. EXPERIMENTAL DESIGN: The effects of AG14361, in comparison with the methylguanine DNA methyltransferase inhibitor, benzylguanine, on temozolomide-induced growth inhibition were investigated in matched pairs of MMR-proficient (HCT-Ch3, A2780, and CP70-ch3) and -deficient (HCT116, CP70, and CP70-ch2) cells. RESULTS: AG14361 enhanced temozolomide activity in all MMR-proficient cells (1.5-3.3-fold) but was more effective in MMR-deficient cells (3.7-5.2-fold potentiation), overcoming temozolomide resistance. In contrast, benzylguanine only increased the efficacy of temozolomide in MMR-proficient cells but was ineffective in MMR-deficient cells. The differential effect of AG14361 in MMR-deficient cells was not attributable to differences in PARP-1 activity or differences in its inhibition by AG14361, nor was it attributable to differences in DNA strand breaks induced by temozolomide plus AG14361. MMR-deficient cells are resistant to cisplatin, but AG14361 did not sensitize any cells to cisplatin. PARP-1 inhibitors potentiate topotecan-induced growth inhibition, but AG14361 did not potentiate topotecan in MMR-deficient cells more than in MMR-proficient cells. CONCLUSIONS: MMR defects are relatively common in sporadic tumors and cancer syndromes. PARP-1 inhibition represents a novel way of selectively targeting such tumors. The underlying mechanism is probably a shift of the cytotoxic locus of temozolomide to N(7)-methylguanine and N(3)-methyladenine, which are repaired by the base excision repair pathway in which PARP-1 actively participates.


Subject(s)
Base Pair Mismatch , Benzodiazepines/pharmacology , DNA Repair , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Enzyme Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Adenine/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Azulenes , Cell Division , Cell Line, Tumor , Cisplatin/pharmacology , DNA Damage , Dose-Response Relationship, Drug , Drug Synergism , Genotype , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Models, Chemical , NAD , Temozolomide
14.
J Natl Cancer Inst ; 96(1): 56-67, 2004 Jan 07.
Article in English | MEDLINE | ID: mdl-14709739

ABSTRACT

BACKGROUND: Poly(ADP-ribose) polymerase-1 (PARP-1) facilitates the repair of DNA strand breaks. Inhibiting PARP-1 increases the cytotoxicity of DNA-damaging chemotherapy and radiation therapy in vitro. Because classical PARP-1 inhibitors have limited clinical utility, we investigated whether AG14361, a novel potent PARP-1 inhibitor (inhibition constant <5 nM), enhances the effects of chemotherapy and radiation therapy in human cancer cell cultures and xenografts. METHODS: The effect of AG14361 on the antitumor activity of the DNA alkylating agent temozolomide, topoisomerase I poisons topotecan or irinotecan, or x-irradiation or gamma-radiation was investigated in human cancer cell lines A549, LoVo, and SW620 by proliferation and survival assays and in xenografts in mice by tumor volume determination. The specificity of AG14361 for PARP-1 was investigated by microarray analysis and by antiproliferation and acute toxicity assays in PARP-1-/- and PARP-1+/+ cells and mice. After intraperitoneal administration, the concentration of AG14361 was determined in mouse plasma and tissues, and its effect on PARP-1 activity was determined in tumor homogenates. All statistical tests were two-sided. RESULTS: AG14361 at 0.4 micro M did not affect cancer cell gene expression or growth, but it did increase the antiproliferative activity of temozolomide (e.g., in LoVo cells by 5.5-fold, 95% confidence interval [CI] = 4.9-fold to 5.9-fold; P =.004) and topotecan (e.g., in LoVo cells by 1.6-fold, 95% CI = 1.3-fold to 1.9-fold; P =.002) and inhibited recovery from potentially lethal gamma-radiation damage in LoVo cells by 73% (95% CI = 48% to 98%). In vivo, nontoxic doses of AG14361 increased the delay of LoVo xenograft growth induced by irinotecan, x-irradiation, or temozolomide by two- to threefold. The combination of AG14361 and temozolomide caused complete regression of SW620 xenograft tumors. AG14361 was retained in xenografts in which PARP-1 activity was inhibited by more than 75% for at least 4 hours. CONCLUSION: AG14361 is, to our knowledge, the first high-potency PARP-1 inhibitor with the specificity and in vivo activity to enhance chemotherapy and radiation therapy of human cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Benzodiazepines/pharmacology , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Radiation-Sensitizing Agents/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azulenes , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/radiotherapy , Dacarbazine/pharmacology , Gamma Rays/therapeutic use , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Mice , Poly(ADP-ribose) Polymerases/metabolism , Temozolomide , Transplantation, Heterologous
15.
Clin Cancer Res ; 9(7): 2711-8, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12855651

ABSTRACT

The nuclear enzyme poly(ADP-ribose) polymerase (PARP-1) facilitates DNA repair, and is, therefore, an attractive target for anticancer chemo- and radio-potentiation. Novel benzimidazole-4-carboxamides (BZ1-6) and tricyclic lactam indoles (TI1-5) with PARP-1 K(i) values of <10 nM have been identified. Whole cell PARP-1 inhibition, intrinsic cell growth inhibition, and chemopotentiation of the cytotoxic agents temozolomide (TM) and topotecan (TP) were evaluated in LoVo human colon carcinoma cells. The acute toxicity of the inhibitors was investigated in PARP-1 null and wild-type mice. Tissue distribution and in vivo chemopotentiation activity was determined in nude mice bearing LoVo xenografts. At a nontoxic concentration (0.4 micro M) the PARP-1 inhibitors potentiated TM-induced growth inhibition 1.0-5.3-fold and TP-induced inhibition from 1.0-2.1-fold. Concentrations of the PARP-1 inhibitors that alone inhibited cell growth by 50% ranged from 8 to 94 micro M. Maximum potentiation of TM activity was achieved at nongrowth inhibitory concentrations (

Subject(s)
Dacarbazine/analogs & derivatives , Enzyme Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Cell Division , Cell Line, Tumor , DNA Repair , Dacarbazine/therapeutic use , Dacarbazine/toxicity , Dose-Response Relationship, Drug , Humans , Indoles/metabolism , Kinetics , Models, Chemical , Poly(ADP-ribose) Polymerases/genetics , Temozolomide , Temperature , Time Factors , Tissue Distribution , Topotecan/therapeutic use , Topotecan/toxicity
16.
J Med Chem ; 45(23): 4961-74, 2002 Nov 07.
Article in English | MEDLINE | ID: mdl-12408707

ABSTRACT

A series of novel compounds have been designed that are potent inhibitors of poly(ADP-ribose) polymerase-1 (PARP-1), and the activity and physical properties have been characterized. The new structural classes, 3,4,5,6-tetrahydro-1H-azepino[5,4,3-cd]indol-6-ones and 3,4-dihydropyrrolo[4,3,2-de]isoquinolin-5-(1H)-ones, have conformationally locked benzamide cores that specifically interact with the PARP-1 protein. The compounds have been evaluated with in vitro cellular assays that measure the ability of the PARP-1 inhibitors to enhance the effect of cytotoxic agents against cancer cell lines.


Subject(s)
Antineoplastic Agents/chemical synthesis , Dacarbazine/analogs & derivatives , Enzyme Inhibitors/chemical synthesis , Indoles/chemical synthesis , Isoquinolines/chemical synthesis , Poly(ADP-ribose) Polymerase Inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Crystallography, X-Ray , Dacarbazine/pharmacology , Drug Design , Drug Screening Assays, Antitumor , Drug Synergism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Indoles/chemistry , Indoles/pharmacology , Isoquinolines/chemistry , Isoquinolines/pharmacology , NAD/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Structure-Activity Relationship , Temozolomide , Topotecan/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...