Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 12(7): 1108-1115, 2021 Jul 08.
Article in English | MEDLINE | ID: mdl-34267880

ABSTRACT

Cyclin-dependent kinase 9 (CDK9) is a serine/threonine kinase involved in the regulation of transcription elongation. An inhibition of CDK9 downregulates a number of short-lived proteins responsible for tumor maintenance and survival, including the antiapoptotic BCL-2 family member MCL-1. As pan-CDK inhibitors under development have faced dosing and toxicity challenges in the clinical setting, we generated selective CDK9 inhibitors that could be amenable to an oral administration. Here, we report the lead optimization of a series of azaindole-based inhibitors. To overcome early challenges with promiscuity and cardiovascular toxicity, carboxylates were introduced into the pharmacophore en route to compounds such as 14 and 16. These CDK9 inhibitors demonstrated a reduced toxicity, adequate pharmacokinetic properties, and a robust in vivo efficacy in mice upon oral dosing.

2.
Front Neurosci ; 12: 889, 2018.
Article in English | MEDLINE | ID: mdl-30546294

ABSTRACT

Obsessive-compulsive disorder (OCD) patients have difficulty in switching between obsessive thought and compulsive behavior, which may be related to the dysfunction of the salience network (SN). However, little is known about the changes in intra- and inter- intrinsic functional connectivity (iFC) of the SN in patients with OCD. In this study, we parceled the SN into 19 subregions and investigated iFC changes for each of these subregions in 40 drug-naïve patients with OCD and 40 healthy controls (HCs) using seed-based functional connectivity resting-state functional magnetic resonance imaging (rs-fMRI). We found that patients with OCD exhibited decreased iFC strength between subregions of the SN, as well as decreased inter-network connectivity between SN and DMN, and ECN. These findings highlight a specific alteration in iFC patterns associated with SN in patients with OCD and provide new insights into the dysfunctional brain organization of the SN in patients with OCD.

3.
Yao Xue Xue Bao ; 48(7): 1053-70, 2013 Jul.
Article in Chinese | MEDLINE | ID: mdl-24133971

ABSTRACT

Chemotherapy remains one of the major tools, along with surgery, radiotherapy, and more recently targeted therapy, in the war against cancer. There have appeared a plethora of highly potent cytotoxic drugs but the poor discriminability between cancerous and healthy cells of these agents limits their broader application in clinical settings. Therapeutic antibodies have emerged as an important class of biological anticancer agents, thanks to their ability in specific binding to tumor-associated antigens. While this important class of biologics can be used as single agents for the treatment of cancer through antibody-dependent cell cytotoxicity (ADCC), their therapeutical efficacy is often limited. Antitumor antibody drug conjugates (ADCs) combine the target-specificity of monoclonal antibody (mAb) and the highly active cell-killing drugs, taking advantages of the best characteristics out of both components. Thus, insufficiency of most naked mAbs in cancer therapy has been circumvented by arming the immunoglobulin with cytotoxic drugs. Here mAbs are used as vehicles to transport potent payloads to tumor cells. ADCs contain three main components: antibody, linker and cytotoxics (also frequently referred as payload). Antibodies can recognize and specifically bind to the tumor-specific antigens, leading to an antibody-assisted internalization, and payload release. While ADC has demonstrated tremendous success, a number of practical challenges limit the broader applications of this new class of anticancer therapy, including inefficient cellular uptake, low cytotoxicity, and off-target effects. This review article aims to cover recent advances in optimizing linkers with increased stability in circulation while allowing efficient payload release within tumor cells. We also attempt to provide some practical strategies in resolving the current challenges in this attractive research area, particularly to those new to the field.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Cytotoxins/therapeutic use , Drug Design , Immunoconjugates/therapeutic use , Neoplasms/drug therapy , Aminobenzoates/pharmacology , Aminobenzoates/therapeutic use , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Cytotoxins/pharmacology , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Maytansine/pharmacology , Maytansine/therapeutic use , Neoplasms/pathology , Oligopeptides/pharmacology , Oligopeptides/therapeutic use
4.
Bioorg Med Chem ; 20(15): 4635-45, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22766219

ABSTRACT

PARP-1, the most abundant member of the PARP superfamily of nuclear enzymes, has emerged as a promising molecular target in the past decade particularly for the treatment of cancer. A number of PARP-1 inhibitors, including veliparab discovered at Abbott, have advanced into different stages of clinical trials. Herein we describe the development of a new tetrahydropyridopyridazinone series of PARP-1 inhibitors. Many compounds in this class, such as 20w, displayed excellent potency against the PARP-1 enzyme with a K(i) value of <1nM and an EC(50) value of 1nM in a C41 whole cell assay. The presence of the NH in the tetrahydropyridyl ring of the tetrahydropyridopyridazinone scaffold improved the pharmacokinetic properties over similar carbon based analogs. Compounds 8c and 20u are orally available, and have demonstrated significant efficacy in a B16 murine xenograft model, potentiating the efficacy of temozolomide (TMZ).


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Neoplasms, Experimental/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Pyridazines/pharmacology , Pyridines/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemical synthesis , Female , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Neoplasms, Experimental/enzymology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Pyridazines/chemical synthesis , Pyridazines/chemistry , Pyridines/chemical synthesis , Pyridines/chemistry , Structure-Activity Relationship , Xenograft Model Antitumor Assays
5.
Clin Exp Med ; 12(3): 135-43, 2012 Sep.
Article in English | MEDLINE | ID: mdl-21874584

ABSTRACT

Gaucher disease is an autosomal recessive lysosomal storage disorder resulting in a deficiency of glucocerebrosidase (GC). Imiglucerase, a recombinant form of GC, has been successfully used in the treatment of Gaucher disease and has been shown to be a good potential candidate for gene therapy. However, its low transduction efficiency and short duration of expression have limited it as a gene therapy strategy. VP22, the herpes simplex virus type I tegument protein, is known to facilitate intercellular protein transport, thus making it a promising tool for improving gene transfer efficiency. To investigate whether the fusion of VP22 to GC could improve its therapeutic efficiency for Gaucher disease, the lentiviral vectors pHIV-GC and pHIV-VP(22)-GC were constructed and confirmed by PCR or RT-PCR. After packaging, the vectors were transduced into human Gaucher II fibroblast cells (GII cells). Flow cytometric analysis revealed that the GC expression rates in lenti-VP(22)-GC-transduced GII cells were higher by comparison than those in lenti-GC-transduced GII cells. A Western blot demonstrated higher levels of GC protein expression in lenti-VP(22)-GC-transduced GII cells. In addition, the long-term expression levels and increased GC activities in lenti-VP(22)-GC-transduced GII cells were also observed. These data implicate that VP22-mediated effects may be useful for enhancing the efficacy of this Gaucher disease treatment.


Subject(s)
Fibroblasts/enzymology , Gaucher Disease/therapy , Gene Transfer Techniques , Genetic Vectors , Glucosylceramidase/metabolism , Viral Structural Proteins/metabolism , Enzyme Activation , Fibroblasts/cytology , Flow Cytometry , Fluorescent Antibody Technique , Gaucher Disease/genetics , Gene Expression Regulation, Enzymologic , Glucosylceramidase/genetics , HEK293 Cells , Herpesvirus 1, Human/genetics , Humans , Lentivirus/genetics , Plasmids/genetics , Plasmids/metabolism , Primary Cell Culture , Transfection , Viral Structural Proteins/genetics
6.
J Med Chem ; 53(8): 3142-53, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20337371

ABSTRACT

We have developed a series of phenylpyrrolidine- and phenylpiperidine-substituted benzimidazole carboxamide poly(ADP-ribose) polymerase (PARP) inhibitors with excellent PARP enzyme potency as well as single-digit nanomolar cellular potency. These efforts led to the identification of (S)-2-(2-fluoro-4-(pyrrolidin-2-yl)phenyl)-1H-benzimidazole-4-carboxamide (22b, A-966492). Compound 22b displayed excellent potency against the PARP-1 enzyme with a K(i) of 1 nM and an EC(50) of 1 nM in a whole cell assay. In addition, 22b is orally bioavailable across multiple species, crosses the blood-brain barrier, and appears to distribute into tumor tissue. It also demonstrated good in vivo efficacy in a B16F10 subcutaneous murine melanoma model in combination with temozolomide and in an MX-1 breast cancer xenograft model both as a single agent and in combination with carboplatin.


Subject(s)
Antineoplastic Agents/chemical synthesis , Benzimidazoles/chemical synthesis , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , BRCA1 Protein/deficiency , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Biological Availability , Blood-Brain Barrier/metabolism , Carboplatin/administration & dosage , Cell Line, Tumor , Crystallography, X-Ray , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Drug Screening Assays, Antitumor , Female , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred C57BL , Mice, SCID , Models, Molecular , Neoplasm Transplantation , Stereoisomerism , Structure-Activity Relationship , Temozolomide , Transplantation, Heterologous
7.
Bioorg Med Chem Lett ; 20(3): 1023-6, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20045315

ABSTRACT

Through conformational restriction of a benzamide by formation of a seven-membered hydrogen-bond with an oxindole carbonyl group, a series of PARP inhibitors was designed for appropriate orientation for binding to the PARP surface. This series of compounds with a 3-oxoisoindoline-4-carboxamide core structure, displayed modest to good activity against PARP-1 in both intrinsic and cellular assays. SAR studies at the lactam nitrogen of the pharmacophore have suggested that a secondary or tertiary amine is important for cellular potency. An X-ray structure of compound 1e bound to the protein confirmed the formation of a seven-membered intramolecular hydrogen bond. Though revealed previously in peptides, this type of seven-membered intramolecular hydrogen bond is rarely observed in small molecules. Largely due to the formation of the intramolecular hydrogen bond, the 3-oxoisoindoline-4-carboxamide core structure appears to be planar in the X-ray structure. An additional hydrogen bond interaction of the piperidine nitrogen to Gly-888 also contributes to the binding affinity of 1e to PARP-1.


Subject(s)
Amides/chemistry , Antineoplastic Agents/chemistry , Drug Discovery/methods , Isoindoles/chemistry , Neoplasms/enzymology , Poly(ADP-ribose) Polymerase Inhibitors , Amides/metabolism , Amides/therapeutic use , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Crystallography, X-Ray , Isoindoles/metabolism , Isoindoles/therapeutic use , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerases/metabolism , Structure-Activity Relationship
8.
Clin Cancer Res ; 15(23): 7277-90, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19934293

ABSTRACT

PURPOSE: ABT-888, currently in phase 2 trials, is a potent oral poly(ADP-ribose) polymerase inhibitor that enhances the activity of multiple DNA-damaging agents, including temozolomide (TMZ). We investigated ABT-888+TMZ combination therapy in multiple xenograft models representing various human tumors having different responses to TMZ. EXPERIMENTAL DESIGN: ABT-888+TMZ efficacy in xenograft tumors implanted in subcutaneous, orthotopic, and metastatic sites was assessed by tumor burden, expression of poly(ADP-ribose) polymer, and O(6)-methylguanine methyltransferase (MGMT). RESULTS: Varying levels of ABT-888+TMZ sensitivity were evident across a broad histologic spectrum of models (55-100% tumor growth inhibition) in B-cell lymphoma, small cell lung carcinoma, non-small cell lung carcinoma, pancreatic, ovarian, breast, and prostate xenografts, including numerous regressions. Combination efficacy in otherwise TMZ nonresponsive tumors suggests that TMZ resistance may be overcome by poly(ADP-ribose) polymerase inhibition. Profound ABT-888+TMZ efficacy was seen in experimental metastases models that acquired resistance to TMZ. Moreover, TMZ resistance was overcome in crossover treatments, indicating that combination therapy may overcome acquired TMZ resistance. Neither tumor MGMT, mismatch repair, nor poly(ADP-ribose) polymer correlated with the degree of sensitivity to ABT-888+TMZ. CONCLUSIONS: Robust ABT-888+TMZ efficacy is observed across a spectrum of tumor types, including orthotopic and metastatic implantation. As many TMZ nonresponsive tumors proved sensitive to ABT-888+TMZ, this novel combination may broaden the clinical use of TMZ beyond melanoma and glioma. Although TMZ resistance may be influenced by MGMT, neither MGMT nor other mechanisms of TMZ resistance (mismatch repair) precluded sensitivity to ABT-888+TMZ. Underlying mechanisms of TMZ resistance in these models are not completely understood but likely involve mechanisms independent of MGMT.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzimidazoles/administration & dosage , Dacarbazine/analogs & derivatives , Animals , Antineoplastic Agents, Alkylating/administration & dosage , DNA Damage , DNA Modification Methylases/metabolism , DNA Repair , DNA Repair Enzymes/metabolism , Dacarbazine/administration & dosage , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Mice , Mice, SCID , Neoplasm Metastasis , Neoplasm Transplantation , Temozolomide , Tumor Suppressor Proteins/metabolism
9.
Mol Cancer Res ; 7(10): 1686-92, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19825992

ABSTRACT

Many established cancer therapies involve DNA-damaging chemotherapy or radiotherapy. Gain of DNA repair capacity of the tumor represents a common mechanism used by cancer cells to survive DNA-damaging therapy. Poly(ADP-ribose) polymerase-1 (PARP-1) is a nuclear enzyme that is activated by DNA damage and plays a critical role in base excision repair. Inhibition of PARP represents an attractive approach for the treatment of cancer. Previously, we have described the discovery and characterization of a potent PARP inhibitor, ABT-888. ABT-888 potentiates the activity of DNA-damaging agents such as temozolomide (TMZ) in a variety of preclinical models. We report here the generation of HCT116 cells resistant to treatment with TMZ and ABT-888 (HCT116R cells). HCT116R cells exhibit decreased H2AX phosphorylation in response to treatment with TMZ and ABT-888 relative to parental HCT116 cells. Microarray and Western blot studies indicate that HCT116R cells have decreased PARP-1 and elevated Rad51 expression levels. HCT116R cells are dependent on Rad51 for proliferation and survival, as shown by inhibition of proliferation and induction of apoptosis upon treatment with Rad51 small interfering RNA. In addition, HCT116R cells are more resistant to radiation than the parental HCT116 cells. Our study suggests that cancer cells upregulate the homologous recombination DNA repair pathway to compensate for the loss of base excision repair, which may account for the observed resistance to treatment with TMZ and ABT-888.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzimidazoles/pharmacology , DNA Repair/drug effects , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Recombination, Genetic/drug effects , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Repair/genetics , Dacarbazine/pharmacology , Down-Regulation/genetics , Histones/drug effects , Histones/genetics , Histones/metabolism , Humans , Phosphorylation/drug effects , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/drug effects , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , RNA, Small Interfering , Rad51 Recombinase/drug effects , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Recombination, Genetic/genetics , Sequence Homology , Temozolomide
10.
J Med Chem ; 52(2): 514-23, 2009 Jan 22.
Article in English | MEDLINE | ID: mdl-19143569

ABSTRACT

We have developed a series of cyclic amine-containing benzimidazole carboxamide PARP inhibitors with a methyl-substituted quaternary center at the point of attachment to the benzimidazole ring system. These compounds exhibit excellent PARP enzyme potency as well as single-digit nanomolar cellular potency. These efforts led to the identification of 3a (2-[(R)-2-methylpyrrolidin-2-yl]-1H-benzimidazole-4-carboxamide, ABT-888), currently in human phase I clinical trials. Compound 3a displayed excellent potency against both the PARP-1 and PARP-2 enzymes with a K(i) of 5 nM and in a C41 whole cell assay with an EC(50) of 2 nM. In addition, 3a is aqueous soluble, orally bioavailable across multiple species, and demonstrated good in vivo efficacy in a B16F10 subcutaneous murine melanoma model in combination with temozolomide (TMZ) and in an MX-1 breast cancer xenograft model in combination with either carboplatin or cyclophosphamide.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Enzyme Inhibitors/pharmacology , Melanoma, Experimental/pathology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacokinetics , Biological Availability , Carboplatin/administration & dosage , Cyclophosphamide/administration & dosage , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Dogs , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Female , Haplorhini , Humans , Magnetic Resonance Spectroscopy , Mice , Mice, SCID , Rats , Temozolomide
11.
Anticancer Res ; 28(5A): 2625-35, 2008.
Article in English | MEDLINE | ID: mdl-19035287

ABSTRACT

ABT-888 is a potent, orally bioavailable PARP-1/2 inhibitor shown to potentiate DNA damaging agents. The ability to potentiate temozolomide (TMZ) and develop a biological marker for PARP inhibition was evaluated in vivo. Doses/schedules that achieve TMZ potentiation in the B16F10 syngeneic melanoma model were utilized to develop an ELISA to detect a pharmacodynamic marker, ADP ribose polymers (pADPr), after ABT 888 treatment. ABT-888 enhanced TMZ antitumor activity, in a dose-proportional manner with no observed toxicity (44-75% tumor growth inhibition vs. TMZ monotherapy), but did not show single agent activity. Extended ABT-888 dosing schedules showed no advantage compared to simultaneous TMZ administration. Efficacy correlated with plasma/tumor drug concentrations. Intratumor drug levels correlated with a dose-proportional/time-dependent reduction in pADPr. Potentiation of TMZ activity by ABT-888 correlated with drug levels and inhibition of PARP activity in vivo. ABT-888 is in Phase 1 trials using a validated ELISA based on the assay developed here to assess pharmacological effect.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzimidazoles/pharmacology , Dacarbazine/analogs & derivatives , Melanoma, Experimental/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacokinetics , Cell Line, Tumor , Dacarbazine/administration & dosage , Dacarbazine/pharmacokinetics , Dacarbazine/pharmacology , Drug Administration Schedule , Drug Synergism , Melanoma, Experimental/enzymology , Melanoma, Experimental/metabolism , Mice , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Temozolomide
12.
Mol Cancer Res ; 6(10): 1621-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18922977

ABSTRACT

Poly(ADP-ribose) polymerase (PARP) senses DNA breaks and facilitates DNA repair via the polyADP-ribosylation of various DNA binding and repair proteins. We explored the mechanism of potentiation of temozolomide cytotoxicity by the PARP inhibitor ABT-888. We showed that cells treated with temozolomide need to be exposed to ABT-888 for at least 17 to 24 hours to achieve maximal cytotoxicity. The extent of cytotoxicity correlates with the level of double-stranded DNA breaks as indicated by gammaH2AX levels. In synchronized cells, damaging DNA with temozolomide in the presence of ABT-888 during the S phase generated high levels of double-stranded breaks, presumably because the single-stranded DNA breaks resulting from the cleavage of the methylated nucleotides were converted into double-stranded breaks through DNA replication. As a result, treatment of temozolomide and ABT-888 during the S phase leads to higher levels of cytotoxicity. ABT-888 inhibits poly(ADP-ribose) formation in vivo and enhances tumor growth inhibition by temozolomide in multiple models. ABT-888 is well tolerated in animal models. ABT-888 is currently in clinical trials in combination with temozolomide.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Single-Stranded/drug effects , Dacarbazine/analogs & derivatives , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Cell Death/drug effects , Cell Line, Tumor , DNA Repair/drug effects , DNA Replication/drug effects , Dacarbazine/pharmacology , Disease Models, Animal , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Mice , Rats , Temozolomide
13.
Bioorg Med Chem Lett ; 18(14): 3955-8, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18586490

ABSTRACT

Poly(ADP-ribose) polymerases (PARPs) play significant roles in various cellular functions including DNA repair and control of RNA transcription. PARP inhibitors have been demonstrated to potentiate the effect of cytotoxic agents or radiation in a number of animal tumor models. Utilizing a benzimidazole carboxamide scaffold in which the amide forms a key intramolecular hydrogen bond for optimal interaction with the enzyme, we have identified a novel series of PARP inhibitors containing a quaternary methylene-amino substituent at the C-2 position of the benzimidazole. Geminal dimethyl analogs at the methylene-amino substituent were typically more potent than mono-methyl derivatives in both intrinsic and cellular assays. Smaller cycloalkanes such as cyclopropyl or cyclobutyl were tolerated at the quaternary carbon while larger rings were detrimental to potency. In vivo efficacy data in a B16F10 murine flank melanoma model in combination with temozolomide (TMZ) are described for two optimized analogs.


Subject(s)
Antineoplastic Agents/chemical synthesis , Chemistry, Pharmaceutical/instrumentation , Enzyme Inhibitors/chemical synthesis , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Cell Line, Tumor , Chemistry, Pharmaceutical/methods , DNA/chemistry , DNA Repair , Drug Design , Drug Screening Assays, Antitumor , Enzyme Inhibitors/pharmacology , Kinetics , Mice , Neoplasm Transplantation , Transcription, Genetic
14.
Bioorg Med Chem ; 16(14): 6965-75, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18541433

ABSTRACT

We have developed a series of cyclic amine-containing benzimidazole carboxamide poly(ADP-ribose)polymerase (PARP) inhibitors, with good PARP-1 enzyme potency, as well as cellular potency. These efforts led to the identification of a lead preclinical candidate, 10b, 2-(1-propylpiperidin-4-yl)-1H-benzimidazole-4-carboxamide (A-620223). 10b displayed very good potency against both the PARP-1 enzyme with a K(i) of 8nM and in a whole cell assay with an EC(50) of 3nM. 10b is aqueous soluble, orally bioavailable across multiple species, and demonstrated good in vivo efficacy in a B16F10 subcutaneous murine melanoma model in combination with temozolomide (TMZ) and in an MX-1 breast xenograph model in combination with cisplatin.


Subject(s)
Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Breast Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cisplatin/therapeutic use , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Mice , Structure-Activity Relationship , Temozolomide , Transplantation, Heterologous , Xenograft Model Antitumor Assays
15.
Cancer Biol Ther ; 7(6): 934-41, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18376143

ABSTRACT

Majority of chemotherapeutic agents inhibit tumor growth by inducing apoptosis or necrosis. The DNA alkylating agent, N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), kills cells by necrosis through massive production of DNA strand breaks and subsequent over-activation of PARP. Inhibition of PARP, either through PARP1 genetic ablation or through small molecule PARP inhibitors, protected MNNG-induced cell death in certain cell types including MEF and primary cortical cultures. We report here that a potent PARP inhibitor, ABT-888, facilitates the induction of apoptotic cell death in HeLa cells treated with MNNG. Although the release of cytochrome c from mitochondria to cytosol was observed in HeLa cells treated with either MNNG alone or the combination of MNNG and ABT-888 (MNNG/ABT-888), apoptosis is observed only in HeLa cells treated with MNNG/ABT-888. Bcl-2 family proteins regulate the release of cytochrome c. Downregulation of Bax and Bak by their corresponding siRNAs or overexpression of Bcl-xl inhibited the release of cytochrome c from mitochondria to cytosol, and inhibited apoptosis induced by MNNG/ABT-888. Further examination indicates that ATP concentration is greatly reduced in HeLa cells treated with MNNG alone, but not in HeLa cells treated with MNNG/ABT-888. Reduction of ATP concentration by F0F1-ATP synthase inhibitor oligomycin A renders HeLa cells resistant to the apoptosis induction by treatment with MNNG/ABT-888. Unlike in HeLa cells, ABT-888 protected MNNG induced cell death in normal human fibroblasts. Our study provides evidence that PARP activity determines the fate of HeLa cells by regulating the level of ATP after treatment with MNNG.


Subject(s)
DNA Damage , Gene Expression Regulation, Enzymologic , Poly(ADP-ribose) Polymerases/metabolism , Alkylating Agents/pharmacology , Animals , Apoptosis , Benzimidazoles/pharmacology , Cytochromes c/metabolism , Fibroblasts/metabolism , HeLa Cells , Humans , Methylnitronitrosoguanidine/pharmacology , Mice , NAD/chemistry , Oligomycins/pharmacology
16.
J Med Chem ; 50(13): 2990-3003, 2007 Jun 28.
Article in English | MEDLINE | ID: mdl-17523610

ABSTRACT

Compound 7 was identified as a potent (IC50 = 14 nM), selective, and orally bioavailable (F = 70% in mouse) inhibitor of protein kinase B/Akt. While promising efficacy was observed in vivo, this compound showed effects on depolarization of Purkinje fibers in an in vitro assay and CV hypotension in vivo. Guided by an X-ray structure of 7 bound to protein kinase A, which has 80% homology with Akt in the kinase domain, our efforts have focused on structure-activity relationship (SAR) studies of the phenyl moiety, in an attempt to address the cardiovascular liability and further improve the Akt potency. A novel and efficient synthetic route toward diversely substituted phenyl derivatives of 7 was developed utilizing a copper-mediated aziridine ring-opening reaction as the key step. To improve the selectivity of these Akt inhibitors over other protein kinases, a nitrogen atom was incorporated into selected phenyl analogues of 7 at the C-6 position of the methyl indazole scaffold. These modifications resulted in the discovery of inhibitor 37c with greater potency (IC50 = 0.6 nM vs Akt), selectivity, and improved cardiovascular safety profile. The SARs, pharmacokinetic profile, and CV safety of selected Akt inhibitors will be discussed.


Subject(s)
Hypotension/chemically induced , Indazoles/chemical synthesis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrazoles/chemical synthesis , Pyridines/chemical synthesis , Administration, Oral , Animals , Biological Availability , Crystallography, X-Ray , Dogs , Indazoles/adverse effects , Indazoles/pharmacology , Mice , Models, Molecular , Protein Conformation , Purkinje Fibers/drug effects , Purkinje Fibers/physiology , Pyrazoles/adverse effects , Pyrazoles/pharmacology , Pyridines/adverse effects , Pyridines/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship
17.
Clin Cancer Res ; 13(9): 2728-37, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17473206

ABSTRACT

PURPOSE: To evaluate the preclinical pharmacokinetics and antitumor efficacy of a novel orally bioavailable poly(ADP-ribose) polymerase (PARP) inhibitor, ABT-888. EXPERIMENTAL DESIGN: In vitro potency was determined in a PARP-1 and PARP-2 enzyme assay. In vivo efficacy was evaluated in syngeneic and xenograft models in combination with temozolomide, platinums, cyclophosphamide, and ionizing radiation. RESULTS: ABT-888 is a potent inhibitor of both PARP-1 and PARP-2 with K(i)s of 5.2 and 2.9 nmol/L, respectively. The compound has good oral bioavailability and crosses the blood-brain barrier. ABT-888 strongly potentiated temozolomide in the B16F10 s.c. murine melanoma model. PARP inhibition dramatically increased the efficacy of temozolomide at ABT-888 doses as low as 3.1 mg/kg/d and a maximal efficacy achieved at 25 mg/kg/d. In the 9L orthotopic rat glioma model, temozolomide alone exhibited minimal efficacy, whereas ABT-888, when combined with temozolomide, significantly slowed tumor progression. In the MX-1 breast xenograft model (BRCA1 deletion and BRCA2 mutation), ABT-888 potentiated cisplatin, carboplatin, and cyclophosphamide, causing regression of established tumors, whereas with comparable doses of cytotoxic agents alone, only modest tumor inhibition was exhibited. Finally, ABT-888 potentiated radiation (2 Gy/d x 10) in an HCT-116 colon carcinoma model. In each model, ABT-888 did not display single-agent activity. CONCLUSIONS: ABT-888 is a potent inhibitor of PARP, has good oral bioavailability, can cross the blood-brain barrier, and potentiates temozolomide, platinums, cyclophosphamide, and radiation in syngeneic and xenograft tumor models. This broad spectrum of chemopotentiation and radiopotentiation makes this compound an attractive candidate for clinical evaluation.


Subject(s)
Benzimidazoles/administration & dosage , Benzimidazoles/pharmacokinetics , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Administration, Oral , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Biological Availability , Blood-Brain Barrier/metabolism , Cell Line, Tumor , DNA Damage , Disease Models, Animal , Dogs , Drug Synergism , Female , Haplorhini , Humans , Male , Mice , Mice, Inbred Strains , Rats , Rats, Inbred Strains , Xenograft Model Antitumor Assays
18.
Bioorg Med Chem ; 15(6): 2441-52, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17258463

ABSTRACT

Thr-211 is one of three different amino acid residues in the kinase domain of protein kinase B/Akt as compared to protein kinase A (PKA), a closely related analog in the same AGC family. In an attempt to improve the potency and selectivity of our indazole-pyridine series of Akt inhibitors over PKA, efforts have focused on the incorporation of a chemical functionality to interact with the hydroxy group of Thr-211. Several substituents including an oxygen anion, amino, and nitro groups have been introduced at the C-6 position of the indazole scaffold, leading to a significant drop in Akt potency. Incorporation of a nitrogen atom into the phenyl ring at the same position (i.e., 9f) maintained the Akt activity and, in some cases, improved the selectivity over PKA. The structure-activity relationships of the new pyridine-pyrazolopyridine series of Akt inhibitors and their structural features when bound to PKA are also discussed.


Subject(s)
Enzyme Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrazoles/chemical synthesis , Pyridines/chemical synthesis , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Structure-Activity Relationship , Tumor Cells, Cultured/drug effects
19.
Bioorg Med Chem ; 14(20): 6832-46, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16843670

ABSTRACT

A series of heteroaryl-pyridine containing inhibitors of Akt are reported. The synthesis and structure-activity relationships are discussed, leading to the discovery of a indazole-pyridine analogue (K(i)=0.16 nM). These compounds bind in the ATP binding site, are potent, ATP competitive, and reversible inhibitors of Akt activity. No selectivity amongst the Akt isoforms is observed for this analogue, but there is good selectivity against an panel of other kinases. It is least selective for other members of the AGC family of kinases but is nonetheless 40-fold selective for Akt over PKA. The compound shows cellular activity and significantly slows tumor growth in vivo.


Subject(s)
Indazoles/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyridines/chemistry , Molecular Structure , Protein Kinase Inhibitors/chemistry , Stereoisomerism , Structure-Activity Relationship
20.
Bioorg Med Chem Lett ; 16(12): 3150-5, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16603355

ABSTRACT

The structure-activity relationships of a series of isoquinoline-pyridine-based protein kinase B/Akt antagonists have been investigated in an effort to improve the major short-comings of the lead compound 3, including poor pharmacokinetic profiles in several species (e.g., mouse i.v. t(1/2) = 0.3 h, p.o. F = 0%). Chlorination at C-1 position of the isoquinoline improved its pharmacokinetic property in mice (i.v. t(1/2) = 5.0 h, p.o. F = 51%) but resulted in >500-fold drop in potency. In a mouse MiaPaCa-2 xenograft model, an amino analog 10y significantly slowed the tumor growth, however was accompanied by toxicity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Isoquinolines/chemistry , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyridines/chemistry , Pyridines/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Humans , Mice , Molecular Structure , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/chemical synthesis , Structure-Activity Relationship , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...