Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Breast Cancer Res Treat ; 198(3): 487-498, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36853577

ABSTRACT

BACKGROUND: Veliparib is a poly-ADP-ribose polymerase (PARP) inhibitor, and it has clinical activity with every 3 weeks carboplatin and paclitaxel. In breast cancer, weekly paclitaxel is associated with improved overall survival. We aimed to determine the maximum tolerated dose (MTD) and recommended phase 2 dose (RP2D) of veliparib with weekly carboplatin and paclitaxel as well as safety, pharmacokinetics, and preliminary clinical activity in triple negative breast cancer (TNBC). METHODS: Patients with locally advanced/metastatic solid tumors and adequate organ function were eligible. A standard 3 + 3 dose-escalation design was followed by a TNBC expansion cohort. Veliparib doses ranging from 50 to 200 mg orally bid were tested with carboplatin (AUC 2) and paclitaxel (80 mg/m2) given weekly in a 21-day cycle. Adverse events (AE) were evaluated by CTCAE v4.0, and objective response rate (ORR) was determined by RECIST 1.1. RESULTS: Thirty patients were enrolled, of whom 22 had TNBC. Two dose-limiting toxicities were observed. The RP2D was determined to be 150 mg PO bid veliparib with weekly carboplatin and paclitaxel 2 weeks on, 1 week off, based on hematologic toxicity requiring dose reduction in the first 5 cycles of treatment. The most common grade 3/4 AEs included neutropenia, anemia, and thrombocytopenia. PK parameters of veliparib were comparable to single-agent veliparib. In 23 patients with evaluable disease, the ORR was 65%. In 19 patients with TNBC with evaluable disease, the ORR was 63%. CONCLUSION: Veliparib can be safely combined with weekly paclitaxel and carboplatin, and this triplet combination has promising clinical activity.


Subject(s)
Anemia , Breast Neoplasms , Triple Negative Breast Neoplasms , Humans , Female , Carboplatin , Paclitaxel , Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/etiology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Anemia/chemically induced
2.
Contemp Clin Trials ; 85: 105807, 2019 10.
Article in English | MEDLINE | ID: mdl-31260789

ABSTRACT

The primary goal of an exploratory oncology clinical trial is to identify an effective drug for further development. To account for tumor indication selection error, multiple tumor indications are often selected for simultaneous testing in a basket trial. In this article, we propose optimal and minimax two-stage basket trial designs for exploratory clinical trials. Inactive tumor indications are pruned in stage 1 and the active tumor indications are pooled at end of stage 2 to assess overall effectiveness of the test drug. The proposed designs explicitly control the type I and type II error rates with closed-form sample size formula. They can be viewed as a natural extension of Simon's optimal and minimax two-stage designs for single arm trials to multi-arm basket trials. A simulation study shows that the proposed design method has desirable operating characteristics as compared to other commonly used design methods for exploratory basket trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Clinical Trials as Topic/methods , Neoplasms/drug therapy , Humans , Models, Statistical , Research Design , Treatment Outcome
3.
Future Oncol ; 13(4): 307-320, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27739325

ABSTRACT

Veliparib is an orally administered poly(ADP-ribose) polymerase inhibitor that is being studied in Phase I-III clinical trials, including Phase III studies in non-small-cell lung cancer, ovarian cancer and breast cancer. Tumor cells with deleterious BRCA1 or BRCA2 mutations are deficient in homologous recombination DNA repair and are intrinsically sensitive to platinum therapy and poly(ADP-ribose) polymerase inhibitors. We describe herein the design and rationale of a Phase II trial investigating whether the addition of veliparib to temozolomide or carboplatin/paclitaxel provides clinical benefit over carboplatin/paclitaxel with placebo in patients with locally recurrent or metastatic breast cancer harboring a deleterious BRCA1 or BRCA2 germline mutation (Trial registration: EudraCT 2011-002913-12, NCT01506609).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Clinical Protocols , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacokinetics , Breast Neoplasms/pathology , Carboplatin/administration & dosage , Carboplatin/pharmacokinetics , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacokinetics , Drug Monitoring , Female , Humans , Models, Statistical , Paclitaxel/administration & dosage , Paclitaxel/pharmacokinetics , Research Design , Sample Size , Temozolomide
4.
Cancer Chemother Pharmacol ; 74(3): 583-91, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25053388

ABSTRACT

PURPOSE: A phase 1 study was conducted to evaluate the bioavailability and food effect of a new veliparib formulation in subjects with solid tumors. METHODS: Subjects (planned: Stage I, N = 20; Stage II, N = 16) received four regimens of a single oral dose of veliparib utilizing a group-sequential design. Subjects were administered single doses of 40 mg veliparib supplied as four 10 mg current formulation, four 10 mg new formulation and one 40 mg new formulation under fasting conditions and under non-fasting conditions. Serial blood samples were collected for the determination of veliparib pharmacokinetics. At the end of Stage I, the relative bioavailability between each pair of regimens was assessed by a two one-sided tests procedure from the analyses of the natural logarithms of C(max) and AUC. A 92.7 % confidence interval within the 0.80-1.25 range between each regimen pair determined bioequivalence. RESULTS: Four 10 mg current formulation capsules, four 10 mg new formulation and one 40 mg new formulation were bioequivalent with respect to C(max) and AUC under fasting conditions. The administration of a high-fat meal did not have a significant effect on AUC and only caused a slight decrease in veliparib C(max) (17 %) and a delay of approximately 1 h in T(max). CONCLUSIONS: The 40 mg new capsule was bioequivalent to currently used formulation. Food had no effect on the extent of veliparib absorption and only a small (17 %) decrease in peak exposure of veliparib.


Subject(s)
Benzimidazoles/pharmacokinetics , Diet , Neoplasms/drug therapy , Administration, Oral , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Benzimidazoles/administration & dosage , Biological Availability , Capsules , Fasting , Female , Humans , Male , Middle Aged , Therapeutic Equivalency
5.
Invest New Drugs ; 32(5): 904-12, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24764124

ABSTRACT

Androgen receptor-mediated transcription is directly coupled with the induction of DNA damage, and castration-resistant tumor cells exhibit increased activity of poly (ADP-ribose) polymerase (PARP)-1, a DNA repair enzyme. This study assessed the efficacy and safety of low dose oral PARP inhibitor veliparib (ABT-888) and temozolomide (TMZ) in docetaxel-pretreated patients with metastatic castration-resistant prostate cancer (mCRPC) in a single-arm, open-label, pilot study. Patients with mCRPC progressing on at least one docetaxel-based therapy and prostate specific antigen (PSA) ≥ 2 ng/mL were treated with veliparib 40 mg twice daily on days 1-7 and TMZ once daily (150 mg/m(2)/day cycle 1; if well tolerated then 200 mg/m(2)/day cycle 2 onwards) on days 1-5 q28 days. Patients received 2 (median) treatment cycles (range, 1-9). The primary endpoint was confirmed PSA response rate (decline ≥ 30 %). Twenty-six eligible patients were enrolled, 25 evaluable for PSA response. Median baseline PSA was 170 ng/mL. Two patients had a confirmed PSA response (8.0 %; 95 % CI: 1.0-26.0), 13 stable PSA, and 10 PSA progression. The median progression-free survival was 9 weeks (95 % CI: 7.9-17) and median overall survival 39.6 weeks (95 % CI: 26.6-not estimable). The most frequent treatment-emergent adverse events (AEs) were thrombocytopenia (77 %), anemia (69 %), fatigue (50 %), neutropenia (42 %), nausea (38 %), and constipation (23 %). Grade 3/4 AEs occurring in > 10 % of patients were thrombocytopenia (23 %) and anemia (15 %). Veliparib and TMZ combination was well tolerated but with modest activity. Biomarker analysis supported the proof of concept that this combination has some antitumor activity in mCRPC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzimidazoles/administration & dosage , Benzimidazoles/adverse effects , DNA Repair/drug effects , Dacarbazine/administration & dosage , Dacarbazine/adverse effects , Dacarbazine/analogs & derivatives , Humans , Kallikreins/blood , Male , Middle Aged , Pilot Projects , Poly(ADP-ribose) Polymerase Inhibitors , Prostate-Specific Antigen/blood , Prostatic Neoplasms, Castration-Resistant/blood , Temozolomide , Treatment Outcome
6.
Clin Pharmacokinet ; 53(5): 479-88, 2014 May.
Article in English | MEDLINE | ID: mdl-24452810

ABSTRACT

BACKGROUND AND OBJECTIVE: Veliparib (ABT-888) is a potent oral inhibitor of Poly(ADP-ribose) polymerase enzyme that is currently in development for the treatment of non-hematologic and hematologic malignancies. This analysis characterizes the population pharmacokinetics of veliparib, including developing a structural pharmacokinetic model and testing patient demographics and covariates for potential influence on veliparib pharmacokinetics in patients with non-hematologic malignancies. METHODS: The analysis dataset included 3,542 veliparib concentration values from 325 patients with non-hematologic malignancies enrolled in three phase I and one phase II studies. Population pharmacokinetic modeling was performed using NONMEM. The likelihood ratio test was used for comparison of nested models, and visual predictive check was employed for model qualification. Covariates tested included body size measures, creatinine clearance (CLCR), formulation, age, sex, race, liver function tests, and coadministration with temozolomide. RESULTS: A one-compartment model with first-order absorption and elimination adequately described veliparib pharmacokinetics. The final model included fixed effects for CLCR on veliparib oral clearance (CL/F) and lean body mass (LBM) on volume of distribution (V d/F). CL/F and V d/F were 20.9 L/h (for a CLCR of 100 mL/min) and 173 L (for an LBM of 56 kg), respectively. CONCLUSION: Only LBM and CLCR were found to be determinants of veliparib V d/F and CL/F, respectively. Dosage adjustments of veliparib on the basis of body size, age, sex, race, liver function, and temozolomide coadministration are not necessary in patients with non-hematologic malignancies. This is the first study to characterize the population pharmacokinetics of veliparib, and the developed model will be used to conduct simulations and evaluate veliparib exposure-response relationships.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Benzimidazoles/pharmacokinetics , Models, Biological , Neoplasms/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/blood , Benzimidazoles/blood , Cross-Over Studies , Double-Blind Method , Female , Humans , Male , Middle Aged , Young Adult
7.
Bioorg Med Chem Lett ; 22(24): 7615-22, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23103095

ABSTRACT

A high throughput screening (HTS) hit, 1 (Plk1 K(i)=2.2 µM) was optimized and evaluated for the enzymatic inhibition of Plk-1 kinase. Molecular modeling suggested the importance of adding a hydrophobic aromatic amine side chain in order to improve the potency by a classic kinase H-donor-acceptor binding mode. Extensive SAR studies led to the discovery of 49 (Plk1 K(i)=5 nM; EC(50)=1.05 µM), which demonstrated moderate efficacy at 100 mpk in a MiaPaCa tumor model, with no overt toxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , High-Throughput Screening Assays , Neoplasms, Experimental/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mice , Models, Molecular , Molecular Structure , Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Polo-Like Kinase 1
8.
J Peripher Nerv Syst ; 17(3): 324-30, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22971094

ABSTRACT

Chemotherapy-induced peripheral neuropathy (CIPN) is a major toxicity of chemotherapy treatment for which no therapy is approved. Poly(ADP-ribose) polymerase (PARP)1/2 are nuclear enzymes activated upon DNA damage, and PARP1/2 inhibition provides resistance against DNA damage. A role for PARP inhibition in sensory neurotransmission has also been established. PARP inhibitors attenuate pain-like behaviors and neuropathy-associated decreased peripheral nerve function in diabetic models. The hypothesis tested was that PARP inhibition protects against painful neuropathy. The objective of this study was to investigate whether the novel, selective PARP1/2 inhibitors (ABT-888 and related analogues) would attenuate development of mechanical allodynia in vincristine-treated rats. PARP inhibitors were dosed for 2 days, and then co-administered with vincristine for 12 days. Mechanical allodynia was observed in rats treated with vincristine. PARP1/2 inhibition significantly attenuated development of mechanical allodynia and reduced poly ADP-ribose (PAR) activation in rat skin. The data presented here show that PARP inhibition attenuates vincristine-induced mechanical allodynia in rats, and supports that PARP inhibition may represent a novel therapeutic approach for CIPN.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Benzimidazoles/therapeutic use , Neuralgia/chemically induced , Neuralgia/prevention & control , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Benzimidazoles/pharmacology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Male , Neuralgia/enzymology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Rats , Rats, Sprague-Dawley
9.
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
10.
J Biol Chem ; 286(45): 38960-8, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21890637

ABSTRACT

The ubiquitin/proteasome pathway plays critical roles in virtually all aspects of cell biology. Enzymes of the ubiquitin pathway add (ligases) or remove (deubiquitinases) ubiquitin tags to or from their target proteins in a selective fashion. USP2a is a member of a subfamily of deubiquitinases, called ubiquitin-specific cysteine proteases (USPs). Although USP2a has been reported to be a bona fide oncogene that regulates the stability of MDM2, MDMX, and FAS, it is likely that there are other unidentified substrates for USP2a. In this study, we show that USP2a mediates mitotic progression by regulating the stability of Aurora-A. Through cell-based screening of a USP siRNA library, we discovered that knockdown of USP2a reduced the protein levels of Aurora-A. USP2a interacts with Aurora-A directly in vitro and in vivo. In addition, Aurora-A is a substrate for USP2a in vitro and in vivo. Our study provides a novel mechanism for the role of USP2a in mediating the stability of Aurora-A.


Subject(s)
Endopeptidases/metabolism , Mitosis/physiology , Protein Serine-Threonine Kinases/metabolism , Ubiquitin/metabolism , Aurora Kinases , Endopeptidases/chemistry , Endopeptidases/genetics , Enzyme Stability/physiology , Gene Knockdown Techniques , HeLa Cells , Humans , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Ubiquitin/genetics , Ubiquitin Thiolesterase
11.
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
12.
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
13.
Cancer Chemother Pharmacol ; 65(3): 419-25, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19526240

ABSTRACT

PURPOSE: ABT-888 inhibits poly(ADP-ribose) polymerase (PARP) and may enhance the efficacy of chemotherapy and radiation in CNS tumors. We studied the plasma and cerebrospinal fluid (CSF) pharmacokinetics (PK) of ABT-888 in a non-human primate (NHP) model that is highly predictive of human CSF penetration. METHODS: ABT-888, 5 mg/kg, was administered orally to three NHPs. Serial blood and CSF samples were obtained. Plasma and CSF concentrations of ABT-888 were measured using LC/MS/MS, and the resulting concentration versus time data were evaluated using non-compartmental and compartmental PK methods. RESULTS: The CSF penetration of ABT-888 was 57+/-7% (mean+/-SD). The peak ABT-888 concentration in the plasma was 0.62+/-0.18 microM. Plasma and CSF AUC0-infinity were 3.7+/-1.7 and 2.1+/-0.8 microM h. PARP inhibition in peripheral blood mononuclear cells was evident 2 h after ABT-888 administration. CONCLUSION: The CSF penetration of ABT-888 after oral administration was 57%. Plasma and CSF concentrations were in the range that has been shown to inhibit PARP activity in vivo in humans.


Subject(s)
Benzimidazoles/blood , Benzimidazoles/cerebrospinal fluid , Administration, Oral , Animals , Area Under Curve , Benzimidazoles/pharmacokinetics , Humans , Leukocytes, Mononuclear/enzymology , Macaca mulatta , Male , Metabolic Clearance Rate , Molecular Structure , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Time Factors
14.
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
15.
J Med Chem ; 52(21): 6803-13, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19888760

ABSTRACT

Small molecule inhibitors of PARP-1 have been pursued by various organizations as potential therapeutic agents either capable of sensitizing cytotoxic treatments or acting as stand-alone agents to combat cancer. As one of the strategies to expand our portfolio of PARP-1 inhibitors, we pursued unsaturated heterocycles to replace the saturated cyclic amine derivatives appended to the benzimidazole core. Not only did a variety of these new generation compounds maintain high enzymatic potency, many of them also displayed robust cellular activity. For example, the enzymatic IC(50) and cellular EC(50) values were as low as 1 nM or below. Compounds 24 (EC(50) = 3.7 nM) and 44 (EC(50) = 7.8 nM), featuring an oxadiazole and a pyridine moiety, respectively, demonstrated balanced potency and PK profiles. In addition, these two molecules exhibited potent oral in vivo efficacy in potentiating the cytotoxic agent temozolomide in a B16F10 murine melanoma model.


Subject(s)
Antineoplastic Agents/chemical synthesis , Benzimidazoles/chemical synthesis , Oxadiazoles/chemical synthesis , Poly(ADP-ribose) Polymerase Inhibitors , Pyridines/chemical synthesis , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Alkylating , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , Biological Availability , Cell Line, Tumor , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Drug Synergism , Female , Humans , Male , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Oxadiazoles/pharmacokinetics , Oxadiazoles/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Pyridines/pharmacokinetics , Pyridines/pharmacology , Structure-Activity Relationship , Temozolomide , Transplantation, Heterologous
16.
Cancer Biol Ther ; 8(21): 2004-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19823047

ABSTRACT

PURPOSE: Targeting the poly (ADP-ribose) polymerase (PARP) pathway for cancer treatment has been an active area of pre-clinical and clinical research. We aimed to determine whether the PARP inhibitor ABT-888 hits its therapeutic target in tumors by immunohistochemistry during a Phase 0 trial conducted at the National Cancer Institute. EXPERIMENTAL DESIGN: The expression of poly (ADP-ribose) (PAR) and full size PARP-1 were quantitatively examined by immunohistochemistry in paraffin-embedded tumor biopsies at baseline and 3-24 h after a single oral dose (25 or 50 mg) of ABT-888. RESULTS: Baseline PAR levels were moderate to high in three patients with non-Hodgkin lymphomas, and one each with small cell lung cancer, squamous cell carcinoma of the tongue and melanoma; low in two patients with cutaneous T-cell lymphoma and one with adenocarcinoma of external ear canal. A significant decrease in PAR (median decrease 30.2, range -13.1 to -69.8) was achieved after drug administration (n = 6 pairs; p = 0.03), whereas an increase in PARP-1 expression was observed in five of the six tumors. This resulted in a decrease in the ratio of PAR to PARP-1 in tumor biopsies (median -6.76, range -0.41 to -22.59; p = 0.03). CONCLUSIONS: ABT-888 hits its therapeutic target by significantly reducing PAR levels and the ratio of PAR to PARP-1 in human tumor cells detected by immunohistochemistry. Baseline tumor PAR levels vary considerably among patients who entered this phase 0 study. This underscores a need to investigate baseline PAR levels in association with response in future preclinical and clinical studies.


Subject(s)
Benzimidazoles/pharmacology , Enzyme Inhibitors/pharmacology , Lymphoma/drug therapy , Neoplasms/drug therapy , Neoplasms/enzymology , Poly(ADP-ribose) Polymerase Inhibitors , Blotting, Western , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Immunohistochemistry , Lymphoma/enzymology , Lymphoma/pathology , Neoplasms/pathology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/biosynthesis
17.
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
18.
Bioorg Med Chem Lett ; 19(15): 4050-4, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19553114

ABSTRACT

Based on screening hit 1, a series of tricyclic quinoxalinones have been designed and evaluated for inhibition of PARP-1. Substitutions at the 7- and 8-positions of the quinoxalinone ring led to a number of compounds with good enzymatic and cellular potency. The tricyclic quinoxalinone class is sensitive to modifications of both the amine substituent and the tricyclic core. The synthesis and structure-activity relationship studies are presented.


Subject(s)
Chemistry, Pharmaceutical/methods , Poly(ADP-ribose) Polymerase Inhibitors , Quinoxalines/chemistry , Quinoxalines/chemical synthesis , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis , Cell Nucleus/metabolism , DNA Repair , Drug Design , Drug Screening Assays, Antitumor , Humans , Kinetics , Models, Molecular , Molecular Conformation , Niacinamide/chemistry , Structure-Activity Relationship
19.
J Clin Oncol ; 27(16): 2705-11, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19364967

ABSTRACT

PURPOSE: We conducted the first phase 0 clinical trial in oncology of a therapeutic agent under the Exploratory Investigational New Drug Guidance of the US Food and Drug Administration. It was a first-in-human study of the poly (ADP-ribose) polymerase (PARP) inhibitor ABT-888 in patients with advanced malignancies. PATIENTS AND METHODS: ABT-888 was administered as a single oral dose of 10, 25, or 50 mg to determine the dose range and time course over which ABT-888 inhibits PARP activity in tumor samples and peripheral blood mononuclear cells, and to evaluate ABT-888 pharmacokinetics. Blood samples and tumor biopsies were obtained pre- and postdrug administration for evaluation of PARP activity and pharmacokinetics. A novel statistical approach was developed and utilized to study pharmacodynamic modulation as the primary end point for trials of limited sample size. RESULTS: Thirteen patients with advanced malignancies received the study drug; nine patients underwent paired tumor biopsies. ABT-888 demonstrated good oral bioavailability and was well tolerated. Statistically significant inhibition of poly (ADP-ribose) levels was observed in tumor biopsies and peripheral blood mononuclear cells at the 25-mg and 50-mg dose levels. CONCLUSION: Within 5 months of study activation, we obtained pivotal biochemical and pharmacokinetic data that have guided the design of subsequent phase I trials of ABT-888 in combination with DNA-damaging agents. In addition to accelerating the development of ABT-888, the rapid conclusion of this trial demonstrates the feasibility of conducting proof-of-principle phase 0 trials as part of an alternative paradigm for early drug development in oncology.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Enzyme Inhibitors/therapeutic use , Leukocytes, Mononuclear/drug effects , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Administration, Oral , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacokinetics , Biological Availability , Biopsy , Clinical Trials, Phase I as Topic , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Feasibility Studies , Female , Humans , Leukocytes, Mononuclear/enzymology , Male , Middle Aged , Neoplasms/enzymology , Neoplasms/pathology , Research Design , Treatment Outcome
20.
Mol Cancer Ther ; 8(2): 386-93, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19208828

ABSTRACT

A commonly activated signaling cascade in many human malignancies, including glioblastoma multiforme, is the Akt pathway. This pathway can be activated via numerous upstream alterations including genomic amplification of epidermal growth factor receptor, PTEN deletion, or PIK3CA mutations. In this study, we screened phosphatidylinositol 3-kinase/Akt small-molecule inhibitors in an isogenic cell culture system with an activated Akt pathway secondary to a PIK3CA mutation. One small molecule, A-443654, showed the greatest selective inhibition of cells with the mutant phenotype. Based on these findings, this inhibitor was screened in vitro against a panel of glioblastoma multiforme cell lines. All cell lines tested were sensitive to A-443654 with a mean IC(50) of approximately 150 nmol/L. An analogue of A-443654, methylated at a region that blocks Akt binding, was on average 36-fold less active. Caspase assays and dual flow cytometric analysis showed an apoptotic mechanism of cell death. A-443654 was further tested in a rat intracranial model of glioblastoma multiforme. Animals treated intracranially with polymers containing A-443654 had significantly extended survival compared with control animals; animals survived 79% and 43% longer than controls when A-443654-containing polymers were implanted simultaneously or in a delayed fashion, respectively. This small molecule also inhibited glioblastoma multiforme stem-like cells with similar efficacy compared with traditionally cultured glioblastoma multiforme cell lines. These results suggest that local delivery of an Akt small-molecule inhibitor is effective against experimental intracranial glioma, with no observed resistance to glioblastoma multiforme cells grown in stem cell conditions.


Subject(s)
Glioblastoma/enzymology , Glioblastoma/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Central Nervous System/drug effects , Class I Phosphatidylinositol 3-Kinases , Enzyme Activation/drug effects , Female , Glioblastoma/drug therapy , Humans , Indazoles/pharmacology , Indazoles/toxicity , Indoles/pharmacology , Indoles/toxicity , Mutation/genetics , Neoplastic Stem Cells/enzymology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...