135 results on '"Vincent L, Giranda"'
Search Results
2. Supplementary Data from Acquired Resistance to Combination Treatment with Temozolomide and ABT-888 Is Mediated by Both Base Excision Repair and Homologous Recombination DNA Repair Pathways
- Author
-
Alexander R. Shoemaker, Eric F. Johnson, Joel Leverson, Yan Luo, Vincent L. Giranda, Saul Rosenberg, Thomas Penning, Gui-dong Zhu, Joann Palma, Loren Lasko, Lisa Roberts, Thomas McGonigal, Gang Wang, Dimitri Semizarov, Yan Shi, Mark Anderson, Edward K. Han, and Xuesong Liu
- Abstract
Supplementary Data from Acquired Resistance to Combination Treatment with Temozolomide and ABT-888 Is Mediated by Both Base Excision Repair and Homologous Recombination DNA Repair Pathways
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Data from ABT-888 Confers Broad In vivo Activity in Combination with Temozolomide in Diverse Tumors
- Author
-
Cherrie K. Donawho, David J. Frost, Saul H. Rosenberg, Vincent L. Giranda, Thomas D. Penning, Gui-Dong Zhu, Loren Lasko, Yan Shi, Xuesong Liu, Amanda Niquette, Gail Bukofzer, Paul A. Ellis, Debra Montgomery, Luis E. Rodriguez, Yi-Chun Wang, and Joann P. Palma
- Abstract
Supplementary Data from ABT-888 Confers Broad In vivo Activity in Combination with Temozolomide in Diverse Tumors
- Published
- 2023
- Full Text
- View/download PDF
4. Data from ABT-888 Confers Broad In vivo Activity in Combination with Temozolomide in Diverse Tumors
- Author
-
Cherrie K. Donawho, David J. Frost, Saul H. Rosenberg, Vincent L. Giranda, Thomas D. Penning, Gui-Dong Zhu, Loren Lasko, Yan Shi, Xuesong Liu, Amanda Niquette, Gail Bukofzer, Paul A. Ellis, Debra Montgomery, Luis E. Rodriguez, Yi-Chun Wang, and Joann P. Palma
- 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 O6-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.(Clin Cancer Res 2009;15(23):7277–90)
- Published
- 2023
- Full Text
- View/download PDF
5. 4 Supplementary Figures from Small Interfering RNA–Mediated Polo-Like Kinase 1 Depletion Preferentially Reduces the Survival of p53-Defective, Oncogenic Transformed Cells and Inhibits Tumor Growth in Animals
- Author
-
Yan Luo, Vincent L. Giranda, Daniel Albert, Joel D. Leverson, Paul Tapang, and Ran Guan
- Abstract
4 Supplementary Figures from Small Interfering RNA–Mediated Polo-Like Kinase 1 Depletion Preferentially Reduces the Survival of p53-Defective, Oncogenic Transformed Cells and Inhibits Tumor Growth in Animals
- Published
- 2023
- Full Text
- View/download PDF
6. Data from Small Interfering RNA–Mediated Polo-Like Kinase 1 Depletion Preferentially Reduces the Survival of p53-Defective, Oncogenic Transformed Cells and Inhibits Tumor Growth in Animals
- Author
-
Yan Luo, Vincent L. Giranda, Daniel Albert, Joel D. Leverson, Paul Tapang, and Ran Guan
- Abstract
Polo-like kinase 1 (Plk1) is required for multiple stages of mitosis and is up-regulated in many human malignancies. We depleted Plk1 expression using small interfering RNA (siRNA) and showed defects in bipolar spindle formation and cytokinesis, growth inhibition, and apoptosis induction in human cancer cell lines. To our surprise, depletion of Plk1 in normal human cells did not result in obvious cell cycle defects, and did not induce significant inhibition of cell growth for at least two cell cycles. In addition, Plk1 siRNA inhibited colony formation in soft agar and tumorigenesis in a HT1080 xenograft model in a dose-dependent manner. Analysis with isogenic pairs of cell lines, differing in p53 status, revealed that Plk1 depletion preferentially induced mitotic arrest, aneuploidy, and reduced cell survival in the p53-defective cell lines. No obvious defects were observed in most p53 wild-type cells during the first few cell cycles. In addition, long-term survival studies revealed that p53 facilitates survival upon Plk1 depletion. Therefore, short-term inhibition of Plk1 can kill tumor cells while allowing normal cells to survive. These data validate the episodic inhibition of Plk1 as a very useful approach for cancer treatment.
- Published
- 2023
- Full Text
- View/download PDF
7. Akt Inhibitor A-443654 Interferes with Mitotic Progression by Regulating Aurora A Kinase Expression
- Author
-
Xuesong Liu, Yan Shi, Keith W. Woods, Paul Hessler, Paul Kroeger, Julie Wilsbacher, Jieyi Wang, Jean Y. Wang, Chunying Li, Qun Li, Saul H. Rosenberg, Vincent L. Giranda, and Yan Luo
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Both Akt and Aurora A kinase have been shown to be important targets for intervention for cancer therapy. We report here that Compound A (A-443654), a specific Akt inhibitor, interferes with mitotic progression and bipolar spindle formation. Compound A induces G2/M accumulation, defects in centrosome separation, and formation of either monopolar arrays or disorganized spindles. On the basis of gene expression array studies, we identified Aurora A as one of the genes regulated transcriptionally by Akt inhibitors including Compound A. Inhibition of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway, either by PI3K inhibitor LY294002 or by Compound A, dramatically inhibits the promoter activity of Aurora A, whereas the mammalian target of rapamycin inhibitor has little effect, suggesting that Akt might be responsible for up-regulating Aurora A for mitotic progression. Further analysis of the Aurora A promoter region indicates that the Ets element but not the Sp1 element is required for Compound A-sensitive transcriptional control of Aurora A. Overexpression of Aurora A in cells treated with Compound A attenuates the mitotic arrest and the defects in bipolar spindle formation induced by Akt inhibition. Our studies suggest that that Akt may promote mitotic progression through the transcriptional regulation of Aurora A.
- Published
- 2008
- Full Text
- View/download PDF
8. Optimal Classes of Chemotherapeutic Agents Sensitized by Specific Small-Molecule Inhibitors of Akt In Vitro and In Vivo
- Author
-
Yan Shi, Xuesong Liu, Edward K. Han, Ran Guan, Alexander R. Shoemaker, Anatol Oleksijew, Keith W. Woods, John P. Fisher, Vered Klinghofer, Loren Lasko, Thomas McGonigal, Qun Li, Saul H. Rosenberg, Vincent L. Giranda, and Yan Luo
- Subjects
Akt ,inhibitors ,chemosensitization ,apoptosis ,synergy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Akt is a serine/threonine kinase that transduces survival signals from survival/growth factors. Deregulation and signal imbalance in cancer cells make them prone to apoptosis. Upregulation or activation of Akt to aid the survival of cancer cells is a common theme in human malignancies. We have developed small-molecule Akt inhibitors that are potent and specific. These Akt inhibitors can inhibit Akt activity and block phosphorylation by Akt on multiple downstream targets in cells. Synergy in apoptosis induction was observed when Akt inhibitors were combined with doxorubicin or camptothecin. Akt inhibitor-induced enhancement of topoisomerase inhibitor cytotoxicity was also evident in long-term cell survival assay. Synergy with paclitaxel in apoptosis induction was evident in cells pretreated with paclitaxel, and enhancement of tumor delay by paclitaxel was demonstrated through cotreatment with Akt inhibitor Compound A (A-443654). Combination with other classes of chemotherapeutic agents did not yield any enhancement of cytotoxicity. These findings provide important guidance in selecting appropriate classes of chemotherapeutic agents for combination with Akt inhibitors in cancer treatment.
- Published
- 2005
- Full Text
- View/download PDF
9. Blocking CHK1 Expression Induces Apoptosis and Abrogates the G2 Checkpoint Mechanism
- Author
-
Yan Luo, Shayna K. Rockow-Magnone, Paul E. Kroeger, Leigh Frost, Zehan Chen, Edward K.-H. Han, Shi-Chung Ng, Robert L. Simmer, and Vincent L. Giranda
- Subjects
Chk1 ,antisense ,ribozyme ,checkpoint ,chemotherapy sensitization ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Checkpoint kinase 1 (Chki) is a checkpoint gene that is activated after DNA damage. It phosphorylates and inactivates the Cdc2 activating phosphatase Cdc25C. This in turn inactivates Cdc2, which leads to G2/M arrest. We report that blocking Chki expression by antisense or ribozymes in mammalian cells induces apoptosis and interferes with the G2/M arrest induced by adriamycin. The Chki inhibitor UCN-01 also blocks the G2 arrest after DNA damage and renders cells more susceptible to adriamycin. These results indicate that Chki is an essential gene for the checkpoint mechanism during normal cell proliferation as well as in the DNA damage response.
- Published
- 2001
- Full Text
- View/download PDF
10. Downregulation of Akt1 Inhibits Anchorage-Independent Cell Growth and Induces Apoptosis in Cancer Cells
- Author
-
Xuesong Liu, Yan Shi, Edward K.-H. Han, Zehan Chen, Saul H. Rosenberg, Vincent L. Giranda, Yan Luo, and Shi-Chung Ng
- Subjects
Akt1 ,apoptosis ,antisense ,oligonucleotide ,cancer ,combination treatment ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The serine/threonine kinases, Akti/PKBα, Akt2/PKBβ, and Akt3/PKBγ, play a critical role in preventing cancer cells from undergoing apoptosis. However, the function of individual Akt isoforms in the tumorigenicity of cancer cells is still not well defined. In the current study, we used an AM antisense oligonucleotide (AS) to specifically downregulate Akti protein in both cancer and normal cells. Our data indicate that AM AS treatment inhibits the ability of MiaPaCa-2, H460, HCT-15, and HT1080 cells to grow in soft agar. The treatment also induces apoptosis in these cancer cells as demonstrated by FRCS analysis and a caspase activity assay. Conversely, Akti AS treatment has little effect on the cell growth and survival of normal human cells including normal human fibroblast (NHF), fibroblast from muscle (FBM), and mammary gland epithelial 184135 cells. In addition, AM AS specifically sensitizes cancer cells to typical chemotherapeutic agents. Thus, Akti is indispensable for maintaining the tumorigenicity of cancer cells. Inhibition of AM may provide a powerful sensitization agent for chemotherapy specifically in cancer cells.
- Published
- 2001
- Full Text
- View/download PDF
11. A phase I/II study of veliparib (ABT-888) with radiation and temozolomide in newly diagnosed diffuse pontine glioma: a Pediatric Brain Tumor Consortium study
- Author
-
Patricia Baxter, Xiao-Nan Li, Adekunle M. Adesina, Lindsey Kilburn, Alberto Broniscer, Ibrahim Quaddoumi, Ira J. Dunkel, P. Ansell, Susan M. Blaney, Arnold C. Paulino, Vincent L. Giranda, Edward R. Smith, Patrick A. Thompson, Jack Su, Maryam Fouladi, Tina Young Poussaint, Arzu Onar-Thomas, and Catherine A. Billups
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pediatric Brain Tumor Consortium ,Veliparib ,Newly diagnosed ,chemistry.chemical_compound ,Pharmacokinetics ,Glioma ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Maculopapular rash ,Temozolomide ,Brain Stem Neoplasms ,Humans ,Child ,business.industry ,Brain Neoplasms ,medicine.disease ,Interim analysis ,chemistry ,Benzimidazoles ,Neurology (clinical) ,medicine.symptom ,business ,Pediatric Neuro-Oncology ,medicine.drug - Abstract
Background A Pediatric Brain Tumor Consortium (PBTC) phase I/II trial of veliparib and radiation followed by veliparib and temozolomide (TMZ) was conducted in children with newly diagnosed diffuse intrinsic pontine glioma (DIPG). The objectives were to: (i) estimate the recommended phase II dose (RP2D) of veliparib with concurrent radiation; (ii) evaluate the pharmacokinetic parameters of veliparib during radiation; (iii) evaluate feasibility of intrapatient TMZ dose escalation; (iv) describe toxicities of protocol therapy; and (v) estimate the overall survival distribution compared with historical series. Methods Veliparib was given Monday through Friday b.i.d. during radiation followed by a 4-week rest. Patients then received veliparib at 25 mg/m2 b.i.d. and TMZ 135 mg/m2 daily for 5 days every 28 days. Intrapatient dose escalation of TMZ was investigated for patients experiencing minimal toxicity. Results Sixty-six patients (65 eligible) were enrolled. The RP2D of veliparib was 65 mg/m2 b.i.d. with radiation. Dose-limiting toxicities during radiation with veliparib therapy included: grade 2 intratumoral hemorrhage (n = 1), grade 3 maculopapular rash (n = 2), and grade 3 nervous system disorder (generalized neurologic deterioration) (n = 1). Intrapatient TMZ dose escalation during maintenance was not tolerated. Following a planned interim analysis, it was concluded that this treatment did not show a survival benefit compared with PBTC historical controls, and accrual was stopped for futility. The 1- and 2-year overall survival rates were 37.2% (SE 7%) and 5.3% (SE 3%), respectively. Conclusion Addition of veliparib to radiation followed by TMZ and veliparib was tolerated but did not improve survival for patients with newly diagnosed DIPG. Trial Registration NCT01514201
- Published
- 2020
12. Abstract S2-05: Efficacy and tolerability of veliparib (V; ABT-888) in combination with carboplatin (C) and paclitaxel (P) vs placebo (Plc)+C/P in patients (pts) with BRCA1 or BRCA2 mutations and metastatic breast cancer: A randomized, phase 2 study
- Author
-
Shannon Puhalla, Agnes Jager, Mark E. Robson, Paul K. Marcom, P Bonnet, SJ Isakoff, Christine K. Ratajczak, M. Campone, Jiang Qian, Alan S. Coates, Véronique Diéras, Melinda L. Telli, Hyo S. Han, M Palácová, Q Qin, Susan M. Domchek, Michael Friedlander, Vincent L. Giranda, D Citrin, I. Bondarenko, Stacie Peacock Shepherd, and Bella Kaufman
- Subjects
Cancer Research ,medicine.medical_specialty ,Chemotherapy ,Veliparib ,business.industry ,medicine.medical_treatment ,Cancer ,Phases of clinical research ,medicine.disease ,Gastroenterology ,Metastatic breast cancer ,Carboplatin ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Oncology ,chemistry ,Tolerability ,030220 oncology & carcinogenesis ,Internal medicine ,PARP inhibitor ,medicine ,030212 general & internal medicine ,business - Abstract
Background: Poly(ADP-ribose) polymerase (PARP) inhibitors block DNA damage repair and may thereby enhance the clinical activity of DNA-damaging chemotherapy. Homologous recombination is defective in BRCA1/2-mutated tumors, leading to more error-prone mechanisms of DNA repair and increased sensitivity to PARP inhibition. V is a potent PARP inhibitor that enhances the antitumor activity of platinum agents in preclinical models. This phase 2 trial (NCT01506609) investigated the safety and efficacy of V+C/P or V+ temozolomide (TMZ) vs Plc+C/P in pts with locally recurrent or metastatic breast cancer harboring a BRCA1 or BRCA2 mutation. Results of the V+C/P and Plc+C/P arms are presented; V+TMZ results will be presented separately. Methods: Pts ≥18 years with histologically confirmed locally recurrent or metastatic breast cancer were randomized 1:1:1 to: 1) V 40 mg BID D1–7+TMZ, 28-D cycle; 2) V 120 mg BID D1–7+C AUC 6, D3 and P 175 mg/m2, D3, 21-D cycle; or 3) Plc BID D1–7+C/P. Key eligibility criteria included deleterious BRCA1/2 mutation, ≤2 prior chemotherapies for metastatic disease, no prior platinum agent, and no CNS metastases. Randomization was stratified by hormone receptor status, prior cytotoxic therapy, and ECOG PS. The primary endpoint was progression-free survival (PFS) per RECIST 1.1 of each V arm vs Plc+C/P by independent review. Primary analysis occurred at the 112th PFS event in the V+C/P and Plc+C/P arms. Overall survival (OS), objective response rate (ORR), tolerability, and quality of life were also evaluated. Results: A total of 196 pts (193 BRCA+ per central lab) were randomized to receive double-blinded V+C/P (n=97) or Plc+C/P (n=99). Baseline demographics and disease characteristics were balanced across all treatment arms. Median study drug exposure was 10 cycles for Plc+C/P and 12 cycles for V+C/P. The V+C/P arm demonstrated numeric improvements for both PFS and OS compared to the Plc+C/P arm; improvement in ORR was statistically significant (Table 1). There was no meaningful increase of toxicity with addition of V. The most common treatment-emergent adverse events (AEs) with Plc+C/P or V+C/P were neutropenia (74%/74%), thrombocytopenia (70%/71%), and nausea (58%/71%). Grade ≥3 AEs in ≥30% of pts were neutropenia (55%/56%) and thrombocytopenia (26%/31%), respectively. There was no difference in the use of G-CSF with addition of V. Significant improvements in fatigue, pain, and insomnia (all P Conclusions: This is the first randomized phase 2 trial of a PARP inhibitor in combination with platinum-based therapy for treatment of BRCA1/2-mutated advanced breast cancer. V+C/P demonstrated significantly higher ORR and symptom improvement compared to Plc+C/P, with nonsignificant trends for improved OS and PFS. Phase 3 trials are ongoing. Table 1Efficacy (ITT population – BRCA mutation)Plc+C/P, n=98V+C/P, n=95HR (95% CI); P valuePFS(mo, 95% CI)12.3 (9.3–14.5)14.1 (11.5–16.2)0.789 (0.536–1.162); 0.231OS (mo, 95% CI)25.0 (18.1–34.8)28.5 (22.4–NR)0.725 (0.468–1.121); 0.148ORR, % (95% CI)61.3 (49.7–71.9)77.8 (66.4–86.7)P=0.027 Citation Format: Han HS, Diéras V, Robson ME, Palácová M, Marcom PK, Jager A, Bondarenko I, Citrin D, Campone M, Telli ML, Domchek SM, Friedlander M, Kaufman B, Ratajczak C, Coates A, Bonnet P, Qin Q, Qian J, Giranda VL, Shepherd SP, Isakoff SJ, Puhalla S. Efficacy and tolerability of veliparib (V; ABT-888) in combination with carboplatin (C) and paclitaxel (P) vs placebo (Plc)+C/P in patients (pts) with BRCA1 or BRCA2 mutations and metastatic breast cancer: A randomized, phase 2 study [abstract]. In: Proceedings of the 2016 San Antonio Breast Cancer Symposium; 2016 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2017;77(4 Suppl):Abstract nr S2-05.
- Published
- 2017
- Full Text
- View/download PDF
13. A randomized Phase II study of veliparib with temozolomide or carboplatin/paclitaxel versus placebo with carboplatin/paclitaxel in BRCA1/2 metastatic breast cancer: design and rationale
- Author
-
Shannon Puhalla, Susan M. Domchek, Melinda L. Telli, Hyo Sook Han, Judy Garber, Vincent L. Giranda, Michael Friedlander, Steven J. Isakoff, Qin Qin, Mark E. Robson, David Maag, Stacie Peacock Shepherd, Bella Kaufman, Eric F. Johnson, and Véronique Diéras
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,endocrine system diseases ,Carboplatin ,chemistry.chemical_compound ,0302 clinical medicine ,Clinical Protocols ,Antineoplastic Combined Chemotherapy Protocols ,skin and connective tissue diseases ,veliparib ,BRCA1 Protein ,General Medicine ,Metastatic breast cancer ,female genital diseases and pregnancy complications ,Dacarbazine ,Paclitaxel ,Research Design ,030220 oncology & carcinogenesis ,PARP inhibitor ,Female ,Drug Monitoring ,medicine.drug ,medicine.medical_specialty ,Clinical Trial Protocol ,Veliparib ,PARP trapping ,Breast Neoplasms ,PARP ,03 medical and health sciences ,breast cancer ,Breast cancer ,Internal medicine ,Temozolomide ,medicine ,Humans ,BRCA2 Protein ,Models, Statistical ,business.industry ,BRCA1 ,medicine.disease ,BRCA2 ,synthetic lethality ,030104 developmental biology ,chemistry ,Sample Size ,Cancer research ,DNA damage ,Benzimidazoles ,Ovarian cancer ,business - 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).
- Published
- 2017
- Full Text
- View/download PDF
14. Rhinoviral Capsid-Binding Inhibitors: Structural Basis for Understanding Rhinoviral Biology and for Drug Design
- Author
-
Guy O. Diana and Vincent L. Giranda
- Subjects
Drug ,Capsid ,media_common.quotation_subject ,Computational biology ,Biology ,media_common - Published
- 2018
- Full Text
- View/download PDF
15. Randomized phase II study evaluating veliparib (ABT-888) with temozolomide in patients with metastatic melanoma
- Author
-
Philip Friedlander, Adil Daud, Grant A. McArthur, F. Jiang, Ming Zhu, Nancy Falotico, Omid Hamid, Yan Luo, Ruth Plummer, Mark R. Middleton, Mark D. McKee, Vincent L. Giranda, Nael M. Mostafa, Brenda Chyla, Jane Qian, and Evelyn McKeegan
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Veliparib ,Phases of clinical research ,Poly(ADP-ribose) Polymerase Inhibitors ,Neutropenia ,Placebo ,Young Adult ,chemistry.chemical_compound ,Double-Blind Method ,Internal medicine ,Temozolomide ,medicine ,Humans ,Neoplasm Metastasis ,Adverse effect ,Antineoplastic Agents, Alkylating ,Melanoma ,Aged ,Neoplasm Staging ,Aged, 80 and over ,Leukopenia ,Brain Neoplasms ,Surrogate endpoint ,business.industry ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Dacarbazine ,Survival Rate ,chemistry ,Benzimidazoles ,Drug Therapy, Combination ,Female ,medicine.symptom ,business ,Follow-Up Studies ,medicine.drug - Abstract
Background Veliparib (ABT-888) is a potent, orally bioavailable, small-molecule inhibitor of the DNA repair enzymes poly ADP-ribose polymerase-1 and -2. Veliparib enhances the efficacy of temozolomide (TMZ) and other cytotoxic agents in preclinical tumor models. Patients and methods In this multicenter, double-blind trial, adults with unresectable stage III or IV metastatic melanoma were randomized 1:1:1 to TMZ plus veliparib 20 or 40 mg, or placebo twice daily. Efficacy end points included progression-free survival (PFS), overall survival (OS), and objective response rate (ORR). Results Patients (N = 346) were randomized between February 2009 and January 2010. Median [95% confidence interval (CI)] PFS was 3.7 (3.0–5.5), 3.6 (1.9–4.1), and 2 (1.9–3.7) months in the 20-mg, 40-mg, and placebo arms, respectively. Median (95% CI) OS was 10.8 (9.0–13.1), 13.6 (11.4–15.9), and 12.9 (9.8–14.3) months, respectively; ORR was 10.3%, 8.7%, and 7.0%. Exploratory analyses showed patients with low ERCC1 expression had longer PFS when TMZ was combined with veliparib. Toxicities were as expected for TMZ. The frequencies of thrombocytopenia, neutropenia, and leukopenia were significantly increased in the veliparib groups. Grade 3 or 4 adverse events, mainly hematologic toxicities, were seen in 55%, 63%, and 41% of patients in the 20-mg, 40-mg, and placebo arms, respectively. Conclusions Median PFS with 20 and 40 mg veliparib almost doubled numerically compared with placebo, but the improvements did not reach statistical significance. OS was not increased with veliparib. Toxicities were similar to TMZ monotherapy, but with increased frequency.
- Published
- 2015
- Full Text
- View/download PDF
16. Veliparib in combination with whole brain radiation therapy in patients with brain metastases: results of a phase 1 study
- Author
-
Anthony Brade, Diane Medina, Ming Zhu, Minesh P. Mehta, Martin Dunbar, F. Wang, Terri Leahy, Hao Xiong, Aruna Turaka, H. Ian Robins, Jane Qian, Lawrence Kleinberg, Ding Wang, Vincent L. Giranda, Nael M. Mostafa, Walter J. Curran, and Kyle D. Holen
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Veliparib ,Nausea ,medicine.medical_treatment ,Administration, Oral ,Phases of clinical research ,Breast Neoplasms ,Poly(ADP-ribose) Polymerase Inhibitors ,chemistry.chemical_compound ,Breast cancer ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,medicine ,Humans ,Survival analysis ,Aged ,Aged, 80 and over ,Dose-Response Relationship, Drug ,Brain Neoplasms ,business.industry ,Middle Aged ,medicine.disease ,Combined Modality Therapy ,Survival Analysis ,Primary tumor ,Surgery ,Radiation therapy ,Treatment Outcome ,Neurology ,chemistry ,PARP inhibitor ,Benzimidazoles ,Female ,Neurology (clinical) ,Cranial Irradiation ,medicine.symptom ,business - Abstract
Veliparib, a potent, oral PARP inhibitor, potentiates the antitumor activity of radiation therapy and crosses the blood-brain barrier. This was a phase 1 dose-escalation study evaluating the safety, and secondarily the antitumor activity of veliparib in combination with whole brain radiation therapy (WBRT) in patients with brain metastases, in order to power future trials. Patients with brain metastases from primary solid tumors were treated with WBRT (30.0 or 37.5 Gy in 10 or 15 fractions) and veliparib (escalating doses of 10-300 mg, orally BID). Safety and tumor response were assessed. Observed survival was compared to predicted survival based on a published nomogram. Eighty-one patients (median age 58 years) were treated. The most common primary tumor types were non-small cell lung (NSCLC; n = 34) and breast cancer (n = 25). The most common AEs deemed possibly related to veliparib (AEs, ≥15 %) were fatigue (30 %), nausea (22 %), and decreased appetite (15 %). Fatigue (5 %), hypokalemia and hyponatremia (3 % each) were the only Grade 3/4 AEs deemed possibly related to veliparib observed in ≥2 patients. Although this was an uncontrolled study, preliminary efficacy results were better than predicted: the median survival time (MST, 95 % CI) for the NSCLC subgroup was 10.0 mo (3.9-13.5) and for the breast cancer subgroup was 7.7 mo (2.8-15.0) compared to a nomogram-model-predicted MST of 3.5 mo (3.3-3.8) and 4.9 mo (4.2-5.5). The addition of veliparib to WBRT did not identify new toxicities when compared to WBRT alone. Based on encouraging safety and preliminary efficacy results, a randomized, controlled phase 2b study is ongoing.
- Published
- 2015
- Full Text
- View/download PDF
17. Clinical Pharmacokinetics and Mass Balance of Veliparib in Combination with Temozolomide in Subjects with Nonhematologic Malignancies
- Author
-
Hao Xiong, Wijith Munasinghe, Silpa Nuthalapati, and Vincent L. Giranda
- Subjects
Drug ,Adult ,Male ,Veliparib ,media_common.quotation_subject ,Phases of clinical research ,Administration, Oral ,Biological Availability ,Pharmacology ,030226 pharmacology & pharmacy ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacokinetics ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Temozolomide ,Humans ,Pharmacology (medical) ,Drug Interactions ,media_common ,Aged ,Dose-Response Relationship, Drug ,business.industry ,Middle Aged ,Phase i study ,Clinical trial ,Orally active ,chemistry ,030220 oncology & carcinogenesis ,Area Under Curve ,Benzimidazoles ,Female ,business ,medicine.drug - Abstract
Veliparib is an orally active potent poly(ADP-ribose) polymerase (PARP) inhibitor currently in phase III clinical trials in solid tumors. This phase I study evaluated the pharmacokinetics and mass balance of veliparib administered alone and in combination with temozolomide, and assessed any potential pharmacokinetic drug-drug interaction between veliparib and temozolomide.This was an open-label, dose-escalation study of veliparib in combination with temozolomide in 42 subjects with nonhematologic malignancies. Veliparib was administered orally at doses ranging from 10 to 80 mg twice daily on days 1-7, and temozolomide was administered orally at 150-200 mg/mMean veliparib maximum observed plasma concentration (CVeliparib is a Biopharmaceutical Classification System (BCS) Class 1 compound, with no less than 90% of the dose absorbed and an oral bioavailability of at least 73%. Veliparib is primarily eliminated by renal excretion. Veliparib exhibited linear pharmacokinetics in the 10-80 mg twice-daily dose range. No pharmacokinetic interaction was observed when veliparib and temozolomide were administered together.NCT00526617.
- Published
- 2017
18. Targeting DNA repair with combination veliparib (ABT-888) and temozolomide in patients with metastatic castration-resistant prostate cancer
- Author
-
Jiang Qian, Maha Hussain, Stacie Peacock Shepherd, Brenda Chyla, Jeremy Cetnar, Evelyn McKeegan, Joshi J. Alumkal, Marion Refici-Buhr, Vincent L. Giranda, Susan F. Slovin, and Michael A. Carducci
- Subjects
Male ,medicine.medical_specialty ,DNA Repair ,Veliparib ,Combination therapy ,Pilot Projects ,Poly(ADP-ribose) Polymerase Inhibitors ,Neutropenia ,Gastroenterology ,Article ,chemistry.chemical_compound ,Prostate cancer ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Temozolomide ,medicine ,Humans ,Pharmacology (medical) ,Aged ,Aged, 80 and over ,Pharmacology ,business.industry ,Middle Aged ,Prostate-Specific Antigen ,medicine.disease ,Surgery ,Dacarbazine ,Prostatic Neoplasms, Castration-Resistant ,Prostate-specific antigen ,Treatment Outcome ,Oncology ,Docetaxel ,chemistry ,PARP inhibitor ,Benzimidazoles ,Kallikreins ,business ,medicine.drug - 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.
- Published
- 2014
- Full Text
- View/download PDF
19. Optimal two-stage designs for exploratory basket trials
- Author
-
Cong Chen, Fang Liu, Eric H. Rubin, Cai Iris Wu, Vincent L. Giranda, and Heng Zhou
- Subjects
Clinical Trials as Topic ,Models, Statistical ,business.industry ,Antineoplastic Agents ,General Medicine ,Minimax ,Machine learning ,computer.software_genre ,Clinical trial ,Treatment Outcome ,Research Design ,Sample size determination ,Neoplasms ,Humans ,Medicine ,Pharmacology (medical) ,Artificial intelligence ,Stage (cooking) ,Multiple tumors ,business ,computer ,Selection (genetic algorithm) ,Type I and type II errors - 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.
- Published
- 2019
- Full Text
- View/download PDF
20. EPT-15A PHASE1/2 CLINICAL TRIAL OF VELIPARIB (ABT-888) AND RADIATION FOLLOWED BY MAINTENANCE THERAPY WITH VELIPARIB AND TEMOZOLOMIDE (TMZ) IN PATIENTS WITH NEWLY DIAGNOSED DIFFUSE INTRINSIC PONTINE GLIOMA (DIPG): A PEDIATRIC BRAIN TUMOR CONSORTIUM INTERIM REPORT OF PHASE II STUDY
- Author
-
Arnold C. Paulino, Tina Young Poussaint, Jack Su, Evelyn McKeegan, Susan M. Blaney, Ibrahim Qaddoumi, James M. Boyett, Patrick A. Thompson, Peter Ansell, Arzu Onar Thomas, Catherine A. Billups, Xiao-Nan Li, Alberto Broniscer, Maryam Fouladi, Lindsay Kilburn, Vincent L. Giranda, Patricia Baxter, and Xia Wan
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Pediatric Brain Tumor Consortium ,Veliparib ,Phases of clinical research ,03 medical and health sciences ,chemistry.chemical_compound ,Abstracts ,0302 clinical medicine ,Maintenance therapy ,Internal medicine ,Glioma ,medicine ,Interim report ,Temozolomide ,business.industry ,medicine.disease ,Clinical trial ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Neurology (clinical) ,business ,medicine.drug - Published
- 2016
21. Hit to Lead optimization of a novel class of squarate-containing polo-like kinases inhibitors
- Author
-
Kent D. Stewart, Stevan W. Djuric, Anil Vasudevan, Jennifer J. Bouska, Zhiren Xia, Vincent L. Giranda, Thomas D. Penning, Eric F. Johnson, Loren M. Lasko, Alexander R. Shoemaker, Qingwei Zhang, Michael J. Mitten, Yan Luo, and Vered Klinghofer
- Subjects
Models, Molecular ,Molecular model ,Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Antineoplastic Agents ,Cell Cycle Proteins ,Polo-like kinase ,Protein Serine-Threonine Kinases ,Biochemistry ,PLK1 ,Mice ,Structure-Activity Relationship ,Proto-Oncogene Proteins ,Drug Discovery ,Animals ,Humans ,Potency ,Protein Kinase Inhibitors ,Molecular Biology ,Cell Proliferation ,chemistry.chemical_classification ,Dose-Response Relationship, Drug ,Molecular Structure ,Kinase ,Organic Chemistry ,Aromatic amine ,Neoplasms, Experimental ,Hit to lead ,Combinatorial chemistry ,High-Throughput Screening Assays ,Enzyme ,chemistry ,Molecular Medicine ,Drug Screening Assays, Antitumor - Abstract
A high throughput screening (HTS) hit, 1 (Plk1 Ki = 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 Ki = 5 nM; EC50 = 1.05 μM), which demonstrated moderate efficacy at 100 mpk in a MiaPaCa tumor model, with no overt toxicity.
- Published
- 2012
- Full Text
- View/download PDF
22. CLIN-ONGOING CLINICAL TRIALS
- Author
-
Albert Lai, James E. Herndon, Charles G. Eberhart, Sarah Milla, Erina Yoritsune, Paula L. Griner, Jaishri O. Blakeley, Masayuki Kanamori, Charles J. Nock, Alva B. Weir, Antonio Omuro, Teiji Tominaga, Leigh Ann Bailey, Nancy Contreras, Sam Ryu, Wolfgang Wick, Kelly Wallen, Xingde Li, Lauren E. Abrey, David H. Harter, Gene H. Barnett, Glenn Stevens, Allan H. Friedman, Gabriele E. Tsung, D.M. Brown, Michael A. Vogelbaum, Ameer Abutaleb, Stefan M. Pfister, Emese Filka, T. Cloughesy, Tulika Ranjan, Andrew B. Lassman, Michael D. Prados, Serena Desideri, Timothy F. Cloughesy, Stuart A. Grossman, Eric C. Holland, Darell D. Bigner, Ryo Nishikawa, Sajeel Chowdhary, Boro Dropulic, Lisa M. DeAngelis, Shinji Kawabata, Frank Saran, Thomas J. Kaley, Warren P. Mason, Elizabeth Hovey, Shaan M. Raza, Patricia Lefferts, Amber E Kerstetter, Roger Henriksson, Cathy Brewer, William J. Garner, Lisa Rogers, Lawrence Kleinberg, Heather J. McCrea, Wenxuan Liang, Mario E. Lacouture, Elliot McVeigh, Toshihiko Kuroiwa, John Simes, Craig Nolan, Mark Rosenthal, Jeffrey H. Wisoff, Paul Rosenblatt, Hillard M. Lazarus, James J. Vredenburgh, Andrew E. Sloan, Hua Fung, Igor T. Gavrilovic, Anna K. Nowak, Olivier Chinot, Richard Schwartz, Helen Wheeler, Stacey Green, Tom Mikkelsen, David Zagzag, Michael C. Bloom, Geneviève Legault, Shin-Ichi Miyatake, Ann Livingstone, Elena Pentsova, Henry S. Friedman, Erin Hartnett, Xiaobu Ye, Katherine B. Peters, Jeffrey C. Allen, Dona Kane, Gregg Shepard, Abhay Sanan, Toshihiro Kumabe, Alfredo Quinones-Hinojosa, Tomo Miyata, Amanda Merkelson, Michael Badruddoja, Kathryn M. Field, Jessica Mavadia, Jill S. Barnholtz-Sloan, Jane S. Reese, Matthias A. Karajannis, Hugo Guerrero-Cazares, Stanton L. Gerson, Mythili Shastry, Jeremy N. Rich, Yukihiko Sonoda, Emmy Ludwig, John Sampson, Christopher L. Brown, John H. Suh, Baldassarre Stea, Heather Embree, Kate Sawkins, John D. Hainsworth, Carmen Kut, Vincent L. Giranda, Phioanh L. Nghiemphu, David T.W. Jones, Howard A. Burris, Cabaret Trial Investigators, Girish Dhall, Lawrence Cher, John A. Boockvar, Ingo K. Mellinghoff, Annick Desjardins, David M. Peereboom, Ryuta Saito, Motomasa Furuse, Jeffrey G. Supko, Yoji Yamashita, Kartik Kesavabhotla, Kent C. Shih, Andrey Korshunov, Samuel T. Chao, Marjorie Pazzi, Jeffrey A. Bacha, Bhardwaj Desai, Kurt Schroeder, Robert H. Miller, Lloyd M. Alderson, Jiefeng Xi, Rajul Shah, Naoko Takebe, Richard M. Green, Alireza Mohammad Mohammadi, Kenneth J. Cohen, Michael Fisher, Naomi E. Rance, and Magalie Hilton
- Subjects
Clinical trial ,Abstracts ,Cancer Research ,medicine.medical_specialty ,Oncology ,business.industry ,medicine ,Neurology (clinical) ,Intensive care medicine ,business - Published
- 2012
- Full Text
- View/download PDF
23. Discovery and SAR of orally efficacious tetrahydropyridopyridazinone PARP inhibitors for the treatment of cancer
- Author
-
Cherrie K. Donawho, Jennifer J. Bouska, Chang Park, Yan Luo, Yan Shi, Xuesong Liu, Eric F. Johnson, Donald J. Osterling, Thomas D. Penning, Paul Ellis, Vincent L. Giranda, Jianchun Gong, Alexander R. Shoemaker, Viraj B. Gandhi, Gui-Dong Zhu, and Amanda M. Olson
- Subjects
Models, Molecular ,Pyridines ,Clinical Biochemistry ,Poly (ADP-Ribose) Polymerase-1 ,Administration, Oral ,Pharmaceutical Science ,Antineoplastic Agents ,Poly(ADP-ribose) Polymerase Inhibitors ,Pharmacology ,Crystallography, X-Ray ,Biochemistry ,Poly (ADP-Ribose) Polymerase Inhibitor ,Mice ,Structure-Activity Relationship ,Drug Discovery ,medicine ,Animals ,Structure–activity relationship ,Potency ,Enzyme Inhibitors ,Molecular Biology ,chemistry.chemical_classification ,Temozolomide ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemistry ,Drug discovery ,Organic Chemistry ,Cancer ,Neoplasms, Experimental ,medicine.disease ,Xenograft Model Antitumor Assays ,Mice, Inbred C57BL ,Pyridazines ,Enzyme ,PARP inhibitor ,Molecular Medicine ,Female ,Poly(ADP-ribose) Polymerases ,medicine.drug - 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
- Published
- 2012
- Full Text
- View/download PDF
24. P3-16-05: A Phase II Trial Expansion Cohort of the PARP Inhibitor Veliparib (ABT888) and Temozolomide in BRCA1/2 Associated Metastatic Breast Cancer
- Author
-
SJ Isakoff, Judy Garber, Leif W. Ellisen, Nadine Tung, EP Winer, Stacie Peacock Shepherd, Beth Overmoyer, Paul E. Goss, J Qian, Rebecca Gelman, Vincent L. Giranda, and Karleen Habin
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,education.field_of_study ,Veliparib ,business.industry ,Population ,Phases of clinical research ,medicine.disease ,Metastatic breast cancer ,Surgery ,chemistry.chemical_compound ,Regimen ,Breast cancer ,chemistry ,Internal medicine ,PARP inhibitor ,Cohort ,medicine ,education ,business - Abstract
Background: Veliparib (ABT-888) is a novel oral inhibitor of Poly (ADP-Ribose) Polymerase (PARP) 1 and 2. Veliparib and temozolomide (TMZ) are synergistic in breast cancer xenograft models. We recently conducted a phase II study of TMZ and veliparib in 41 patients (pts) with metastatic breast cancer (MBC). Activity was observed only in pts with known BRCA1 or BRCA2 deleterious mutations, with a response rate (RR) of 50% (4/8) and clinical benefit rate (CBR) of 62.5%. In order to further evaluate the activity and safety of TMZ and veliparib, we enrolled an expansion cohort of 20 additional patients with BRCA1 or 2 mutations. Methods: We previously conducted a single arm phase II trial of veliparib and TMZ in 41 MBC pts. Eligibility included measurable disease, ≥1 prior MBC therapy and PS ≤ 2. Available archived tumor samples were collected. In this expansion cohort, first line therapy for MBC and prior PARP inihibitor therapy were allowed, and eligible patients were required to have a known deleterious BRCA1 or BRCA2 mutation identified from prior clinical testing. The dose of veliparib in the original cohort was reduced from 40 mg PO BID to 30 mg PO BID. In the expansion cohort all patients received veliparib (30 mg PO BID days 1–7) and TMZ (150mg/m2 PO QD days 1–5) on a 28 day cycle. RECIST response was evaluated every 2 cycles. The primary endpoint was overall response rate. Secondary endpoints included PFS, OS, safety and toxicity. Results: Between June 24, 2010 and Sept 29, 2010, 20 eligible pts (median age 42) were enrolled. Baseline characteristics included: median PS=0 (range 0–2); 9 BRCA1 carriers, 9 BRCA2 carriers, and 2 unknown. 17 pts (85%) received prior adjuvant chemotherapy. 9 patients received a prior platinum chemotherapy. The most common grade 3/4 toxicities included thrombocytopenia and neutropenia. Best response for the 20 patients evaluable at the time of abstract submission includes 3 PR (15%), 6 SD (30%), 11 PD, and a clinical benefit rate of 45%. Combined with the initial cohort of 8 known carriers from the original 41 patients, the total RR is 25% (7/28) and clinical benefit rate of 50% (7 PR, 7 SD). The RR was 40% (6/15) in pts without prior platinum treatment, and 9% (1/11) in pts with prior platinum treatment. The median PFS for the 20 BRCA carriers was 85 days, and among patients with prior platinum treatment compared to no prior platinum, the median PFS was 70 and 179 days, respectively. Three pts remain on study, 1 with a CR for >20 mo. Discussion: We previously demonstrated that veliparib and TMZ is an active combination in BRCA1/2 associated MBC. In this larger expansion cohort of 20 additional patients, the combination continued to show activity, although the response rate was not as robust as previously observed. Differences between the original cohort and the expansion cohort may account for some variation in response, such as prior platinum or PARP inhibitors therapy, number of lines of prior therapy, and the dose of veliparib used (40mg vs 30 mg). These results support further evaluation of this regimen in the BRCA1/2 carrier population, and provide the opportunity to evaluate potential factors that may predict response or resistance to this regimen. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr P3-16-05.
- Published
- 2011
- Full Text
- View/download PDF
25. Discovery and SAR of substituted 3-oxoisoindoline-4-carboxamides as potent inhibitors of poly(ADP-ribose) polymerase (PARP) for the treatment of cancer
- Author
-
Gui-Dong Zhu, Xuesong Liu, Yan Shi, Thomas D. Penning, Viraj B. Gandhi, Chang Park, Vered Klinghofer, Vincent L. Giranda, Yan Luo, and Eric F. Johnson
- Subjects
Tertiary amine ,medicine.drug_class ,Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Antineoplastic Agents ,Carboxamide ,Isoindoles ,Poly(ADP-ribose) Polymerase Inhibitors ,Crystallography, X-Ray ,Biochemistry ,Structure-Activity Relationship ,chemistry.chemical_compound ,Neoplasms ,Drug Discovery ,medicine ,Oxindole ,Molecular Biology ,Chemistry ,Hydrogen bond ,Organic Chemistry ,Amides ,Intramolecular force ,PARP inhibitor ,Lactam ,Molecular Medicine ,Poly(ADP-ribose) Polymerases ,Pharmacophore - 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.
- Published
- 2010
- Full Text
- View/download PDF
26. Immunohistochemical detection of poly(ADP-ribose) polymerase inhibition by ABT-888 in patients with refractory solid tumors and lymphomas
- Author
-
Anthony J. Murgo, Gurmeet Kaur, Dat Nguyen, Sherry X. Yang, Vincent L. Giranda, Joseph E. Tomaszewski, Martin Gutierrez, Shivaani Kummar, Seth M. Steinberg, Alice P. Chen, Larry Rubinstein, and James H. Doroshow
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Lymphoma ,Poly ADP ribose polymerase ,Blotting, Western ,Poly (ADP-Ribose) Polymerase-1 ,Poly(ADP-ribose) Polymerase Inhibitors ,Poly (ADP-Ribose) Polymerase Inhibitor ,Cell Line, Tumor ,Neoplasms ,medicine ,Humans ,Enzyme Inhibitors ,Pharmacology ,business.industry ,Melanoma ,Cutaneous T-cell lymphoma ,Cancer ,medicine.disease ,Immunohistochemistry ,Oncology ,Drug Resistance, Neoplasm ,PARP inhibitor ,Molecular Medicine ,Adenocarcinoma ,Benzimidazoles ,Poly(ADP-ribose) Polymerases ,business - Abstract
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.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.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).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.
- Published
- 2009
- Full Text
- View/download PDF
27. Synthesis and Evaluation of a New Generation of Orally Efficacious Benzimidazole-Based Poly(ADP-ribose) Polymerase-1 (PARP-1) Inhibitors as Anticancer Agents
- Author
-
Xuesong Liu, Vincent L. Giranda, Luis E. Rodriguez, Paul A. Ellis, Donald J. Osterling, Magdalena Przytulinska, David Frost, Thomas D. Penning, Jason Stavropoulos, Cherrie K. Donawho, Yunsong Tong, Eric F. Johnson, Joel D. Leverson, Jennifer J. Bouska, Yan Shi, Patrick A. Marcotte, Amanda M. Olson, Sheela A. Thomas, Nirupama B. Soni, and Yan Luo
- Subjects
Male ,Benzimidazole ,Pyridines ,Poly ADP ribose polymerase ,Transplantation, Heterologous ,Melanoma, Experimental ,Poly (ADP-Ribose) Polymerase-1 ,Administration, Oral ,Biological Availability ,Antineoplastic Agents ,Poly(ADP-ribose) Polymerase Inhibitors ,Chemical synthesis ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,Cell Line, Tumor ,Drug Discovery ,Temozolomide ,Animals ,Humans ,Cytotoxicity ,Antineoplastic Agents, Alkylating ,IC50 ,Oxadiazoles ,biology ,Drug Synergism ,Biological activity ,Small molecule ,Dacarbazine ,Mice, Inbred C57BL ,chemistry ,Biochemistry ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,Benzimidazoles ,Female ,Neoplasm Transplantation - 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.
- Published
- 2009
- Full Text
- View/download PDF
28. An enzyme-linked immunosorbent poly(ADP-ribose) polymerase biomarker assay for clinical trials of PARP inhibitors
- Author
-
Luis E. Rodriguez, Paul A. Ellis, Mary Saltarelli, David J. LeBlond, Vincent L. Giranda, Yan Luo, C Thomas Lin, Xuesong Liu, Cherrie K. Donawho, David Frost, Robert J. Kinders, Yan Shi, Milagros Colon-Lopez, and Joann P. Palma
- Subjects
medicine.drug_class ,DNA damage ,DNA repair ,Poly ADP ribose polymerase ,Melanoma, Experimental ,Biophysics ,Antineoplastic Agents ,Enzyme-Linked Immunosorbent Assay ,Poly(ADP-ribose) Polymerase Inhibitors ,Biology ,Biochemistry ,Poly (ADP-Ribose) Polymerase Inhibitor ,Mice ,In vivo ,Temozolomide ,medicine ,Animals ,Humans ,Enzyme Inhibitors ,Molecular Biology ,Clinical Trials as Topic ,Cell Biology ,Molecular biology ,Dacarbazine ,Disease Models, Animal ,Cancer cell ,PARP inhibitor ,Cancer research ,Benzimidazoles ,Female ,Poly(ADP-ribose) Polymerases ,Biomarkers ,Topoisomerase inhibitor - Abstract
Many established cancer therapies involve DNA-damaging chemotherapy or radiotherapy. The DNA repair capacity of the tumor represents a common mechanism used by cancer cells to survive DNA-damaging therapy. Poly(ADP-ribose) polymerase (PARP) is a nuclear enzyme that is activated by DNA damage and has critical roles in DNA repair. Inhibition of PARP potentiates the activity of DNA-damaging agents such as temozolomide, topoisomerase inhibitors and radiation in both in vitro and in vivo preclinical models. Recently, several PARP inhibitors have entered clinical trials either as single agents or in combination with DNA-damaging chemotherapy. Because PARP inhibitors are not cytotoxic, a biomarker assay is useful to guide the selection of an optimal biological dose. We set out to develop an assay that enables us to detect 50% PAR reduction in human tumors with 80% power in a single-plate assay while assuring no more than a 10% false-positive rate. We have developed and optimized an enzyme-linked immunosorbent assay (ELISA) to measure PARP activity that meets the above-mentioned criterion. This robust assay is able to detect PAR levels of 30-2000 pg/ml in both tumor and peripheral blood monocyte samples. In a B16F10 mouse syngeneic tumor model, PARP inhibitor ABT-888 potentiates the effect of temozolomide in suppressing tumor growth, and PARP activity is greatly reduced by ABT-888 at efficacious doses. In summary, the ELISA assay described here is suitable for biomarker studies in clinical trials of PARP inhibitors.
- Published
- 2008
- Full Text
- View/download PDF
29. Discovery and SAR of 2-(1-propylpiperidin-4-yl)-1H-benzimidazole-4-carboxamide: A potent inhibitor of poly(ADP-ribose) polymerase (PARP) for the treatment of cancer
- Author
-
Viraj B. Gandhi, Kennan C. Marsh, Vered Klinghofer, Elizabeth H. Fry, Eric F. Johnson, David Frost, Amanda M. Olson, Vincent L. Giranda, Jianchun Gong, Chang H. Park, Yan Luo, Jennifer J. Bouska, Wolfgang Wernet, Roland Grandel, Gui-Dong Zhu, W. Lubisch, Saul H. Rosenberg, Sheela A. Thomas, Yan Shi, Cherrie K. Donawho, Xuesong Liu, Thomas D. Penning, and Velitchka Bontcheva-Diaz
- Subjects
Benzimidazole ,medicine.drug_class ,Poly ADP ribose polymerase ,Transplantation, Heterologous ,Clinical Biochemistry ,Melanoma, Experimental ,Pharmaceutical Science ,Breast Neoplasms ,Carboxamide ,Poly(ADP-ribose) Polymerase Inhibitors ,Pharmacology ,Biochemistry ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,In vivo ,Antineoplastic Combined Chemotherapy Protocols ,Drug Discovery ,Temozolomide ,medicine ,Animals ,Humans ,Potency ,Enzyme Inhibitors ,Molecular Biology ,Polymerase ,biology ,Organic Chemistry ,Xenograft Model Antitumor Assays ,Dacarbazine ,chemistry ,Enzyme inhibitor ,PARP inhibitor ,biology.protein ,Molecular Medicine ,Benzimidazoles ,Cisplatin - 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 Ki of 8 nM and in a whole cell assay with an EC50 of 3 nM. 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.
- Published
- 2008
- Full Text
- View/download PDF
30. Phase 0 Trials: An Industry Perspective
- Author
-
Rita Tiehen, Terri L. Leahy, Vincent L. Giranda, Gary Gordon, Helen Eliopoulos, and Robert A. Carr
- Subjects
Cancer Research ,medicine.medical_specialty ,Time Factors ,Drug Industry ,Disease ,Poly(ADP-ribose) Polymerase Inhibitors ,Models, Biological ,Phase (combat) ,Humans ,Medicine ,Enzyme Inhibitors ,Intensive care medicine ,Clinical Trials as Topic ,Clinical Trials, Phase I as Topic ,business.industry ,Mechanism (biology) ,Drug discovery ,Clinical study design ,Perspective (graphical) ,Clinical trial ,Oncology ,Drug development ,Drug Design ,Benzimidazoles ,business ,DNA Damage - Abstract
Worldwide, cancer is a leading cause of morbidity and mortality. An increased understanding of the disease and its process has resulted in a multitude of new targeted therapies. The costs as well as time from drug discovery to market, however, remain staggeringly high and protracted, with the majority of compounds never reaching phase III. The concept of an exploratory or phase 0 trial was introduced as a mechanism to enhance and accelerate the overall process of new oncologic drug development. Performance of a phase 0 study allows researchers to better understand the pharmacokinetic and pharmacodynamic properties of compounds in human subjects before initiation of phase I trials. Data gleaned from a phase 0 trial are beneficial not only in prioritizing promising compounds but also in allowing the modification of phase I study design before initiation. To date, few researchers have taken advantage of the potential benefits of phase 0 trials. This review focuses on the purpose as well as the potential merits of phase 0 trials from the perspective of a pharmaceutical company. The review summarizes the experience of a team of researchers with ABT-888, a novel poly (ADP-ribose) polymerase agent that inhibits an enzyme critical for repairing damage to DNA, which is one of the first compounds to be investigated using the phase 0 clinical trial design.
- Published
- 2008
- Full Text
- View/download PDF
31. Poly (ADP-ribose) polymerase activity regulates apoptosis in HeLa cells after alkylating DNA damage
- Author
-
Gui-Dong Zhu, Vincent L. Giranda, Xuesong Liu, Yan Yan Shi, Yan Luo, Thomas D. Penning, and Xu Luo
- Subjects
Alkylating Agents ,Methylnitronitrosoguanidine ,Cancer Research ,Programmed cell death ,DNA damage ,Poly ADP ribose polymerase ,Apoptosis ,Biology ,Gene Expression Regulation, Enzymologic ,HeLa ,Mice ,PARP1 ,Animals ,Humans ,Pharmacology ,Cytochrome c ,Cytochromes c ,Fibroblasts ,NAD ,biology.organism_classification ,Molecular biology ,Oncology ,PARP inhibitor ,biology.protein ,Molecular Medicine ,Benzimidazoles ,Oligomycins ,Poly(ADP-ribose) Polymerases ,DNA Damage ,HeLa Cells - 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.
- Published
- 2008
- Full Text
- View/download PDF
32. Randomized, Placebo-Controlled, Phase II Study of Veliparib in Combination with Carboplatin and Paclitaxel for Advanced/Metastatic Non-Small Cell Lung Cancer
- Author
-
Caroline Nickner, Sergey Orlov, Laszlo Urban, Hao Xiong, Jiang Qian, C. Michael Jones, Suresh S. Ramalingam, Ulrich Keiholz, Normand Blais, E. Juhasz, Mark D. McKee, Fabrice Barlesi, Peter Ansell, Martin Reck, Vera Gorbunova, Julien Mazieres, Ebenezer A. Kio, Vincent L. Giranda, J. Dziubinski, and Qin Qin
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Veliparib ,Paclitaxel ,medicine.medical_treatment ,Phases of clinical research ,Kaplan-Meier Estimate ,Neutropenia ,Placebo ,Disease-Free Survival ,Carboplatin ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Double-Blind Method ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Lung cancer ,Aged ,Proportional Hazards Models ,Chemotherapy ,business.industry ,Middle Aged ,medicine.disease ,Surgery ,030104 developmental biology ,Treatment Outcome ,chemistry ,030220 oncology & carcinogenesis ,Benzimidazoles ,Female ,business - Abstract
Purpose: PARP plays an important role in DNA repair. Veliparib, a PARP inhibitor, enhances the efficacy of platinum compounds and has been safely combined with carboplatin and paclitaxel. The primary endpoint of this phase II trial determined whether addition of veliparib to carboplatin and paclitaxel improved progression-free survival (PFS) in previously untreated patients with advanced/metastatic non–small cell lung cancer. Experimental Design: Patients were randomized 2:1 to carboplatin and paclitaxel with either veliparib or placebo. Veliparib (120 mg) or placebo was given on days 1 to 7 of each 3-week cycle, with carboplatin (AUC = 6 mg/mL/min) and paclitaxel (200 mg/m2) administered on day 3, for a maximum of 6 cycles. Results: Overall, 158 were included (median age, 63 years; male 68%, squamous histology 48%). Median PFS was 5.8 months in the veliparib group versus 4.2 months in the placebo group [HR, 0.72; 95% confidence interval (CI), 0.45–1.15; P = 0.17)]. Median overall survival (OS) was 11.7 and 9.1 months in the veliparib and placebo groups, respectively (HR, 0.80; 95% CI, 0.54–1.18; P = 0.27). In patients with squamous histology, median PFS (HR, 0.54; 95% CI, 0.26–1.12; P = 0.098) and OS (HR, 0.73; 95% CI, 0.43–1.24; P = 0.24) favored veliparib treatment. Objective response rate was similar between groups (veliparib: 32.4%; placebo: 32.1%), but duration of response favored veliparib treatment (HR, 0.47; 95% CI, 0.16–1.42; P = 0.18). Grade III/IV neutropenia, thrombocytopenia, and anemia were comparable between groups. Conclusions: Veliparib combination with carboplatin and paclitaxel was well-tolerated and demonstrated a favorable trend in PFS and OS versus chemotherapy alone. Patients with squamous histology had the best outcomes with veliparib combination. Clin Cancer Res; 23(8); 1937–44. ©2016 AACR.
- Published
- 2016
33. Design and synthesis of pyridine–pyrazolopyridine-based inhibitors of protein kinase B/Akt
- Author
-
Qun Li, Eric F. Johnson, Viraj B. Gandhi, Ran Guan, Saul H. Rosenberg, Jianchun Gong, Vincent S. Stoll, Keith W. Woods, Yan Luo, Vered Klinghofer, Xuesong Liu, Gui-Dong Zhu, Vincent L. Giranda, and Mulugeta Mamo
- Subjects
Pyridines ,Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Crystallography, X-Ray ,Biochemistry ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,Pyrazolopyridine ,Tumor Cells, Cultured ,Humans ,Structure–activity relationship ,Enzyme Inhibitors ,Protein kinase A ,Protein Kinase Inhibitors ,Molecular Biology ,Protein kinase B ,Serine/threonine-specific protein kinase ,Indazole ,biology ,Organic Chemistry ,Pancreatic Neoplasms ,chemistry ,Protein kinase domain ,Enzyme inhibitor ,biology.protein ,Pyrazoles ,Molecular Medicine ,Proto-Oncogene Proteins c-akt - 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.
- Published
- 2007
- Full Text
- View/download PDF
34. Phase I Study of Veliparib (ABT-888) Combined with Cisplatin and Vinorelbine in Advanced Triple-Negative Breast Cancer and/or BRCA Mutation-Associated Breast Cancer
- Author
-
John A. Thompson, Peggy L. Porter, Jamie Guenthoer, Julie Gralow, Jennifer M. Specht, Hannah M. Linden, Stacie Peacock Shepherd, Brian F. Kiesel, Melissa Griffin, Jan H. Beumer, Brenda F. Kurland, Rosa F. Yeh, Eve T. Rodler, Xiaoyu Chai, Vincent L. Giranda, Vijayakrishna K. Gadi, Elizabeth M. Swisher, Larissa A. Korde, and Sandra Strychor
- Subjects
0301 basic medicine ,Oncology ,Adult ,Cancer Research ,medicine.medical_specialty ,Poly Adenosine Diphosphate Ribose ,Veliparib ,DNA Repair ,Receptor, ErbB-2 ,medicine.medical_treatment ,Ubiquitin-Protein Ligases ,Triple Negative Breast Neoplasms ,Pharmacology ,Neutropenia ,Poly(ADP-ribose) Polymerase Inhibitors ,Vinorelbine ,Vinblastine ,Disease-Free Survival ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Humans ,Triple-negative breast cancer ,Aged ,Cisplatin ,BRCA2 Protein ,Chemotherapy ,business.industry ,BRCA mutation ,Middle Aged ,medicine.disease ,Antineoplastic Agents, Phytogenic ,030104 developmental biology ,chemistry ,Receptors, Estrogen ,030220 oncology & carcinogenesis ,Benzimidazoles ,Female ,business ,Receptors, Progesterone ,medicine.drug - Abstract
Purpose: Cisplatin is synergistic with vinorelbine and the PARP inhibitor veliparib, and has antineoplastic activity in triple-negative breast cancer (TNBC) and BRCA mutation–associated breast cancer. This phase I study assessed veliparib with cisplatin and vinorelbine. Experimental Design: A 3+3 dose-escalation design evaluated veliparib administered twice daily for 14 days with cisplatin (75 mg/m2 day 1) and vinorelbine (25 mg/m2 days 1, 8) every 21 days, for 6 to 10 cycles, followed by veliparib monotherapy. Pharmacokinetics, measurement of poly(ADP-ribose) in peripheral blood mononuclear cells, and preliminary efficacy were assessed. IHC and gene-expression profiling were evaluated as potential predictors of response. Results: Forty-five patients enrolled in nine dose cohorts plus five in an expansion cohort at the highest dose level and recommended phase II dose, 300 mg twice daily. The MTD of veliparib was not reached. Neutropenia (36%), anemia (30%), and thrombocytopenia (12%) were the most common grade 3/4 adverse events. Best overall response for 48 patients was radiologic response with 9-week confirmation for 17 (35%; 2 complete, 15 partial), and stable disease for 21 (44%). Germline BRCA mutation presence versus absence was associated with 6-month progression-free survival [PFS; 10 of 14 (71%) vs. 8 of 27 (30%), mid-P = 0.01]. Median PFS for all 50 patients was 5.5 months (95% confidence interval, 4.1–6.7). Conclusions: Veliparib at 300 mg twice daily combined with cisplatin and vinorelbine is well tolerated with encouraging response rates. A phase II randomized trial is planned to assess veliparib's contribution to cisplatin chemotherapy in metastatic TNBC and BRCA mutation–associated breast cancer. Clin Cancer Res; 22(12); 2855–64. ©2016 AACR.
- Published
- 2015
35. Quantitative Analysis of Anti-apoptotic Function of Akt in Akt1 and Akt2 Double Knock-out Mouse Embryonic Fibroblast Cells under Normal and Stressed Conditions
- Author
-
Morris J. Birnbaum, Tillman Oltersdorf, Yan Luo, Yan Shi, Keqiang Ye, Vincent L. Giranda, Ron De Jong, and Xuesong Liu
- Subjects
Small interfering RNA ,Cell Survival ,AKT1 ,Apoptosis ,AKT2 ,Biology ,Biochemistry ,AKT3 ,Cell Line ,Mice ,Animals ,Phosphorylation ,RNA, Small Interfering ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Mice, Knockout ,Cell Biology ,Fibroblasts ,Flow Cytometry ,Cell biology ,embryonic structures ,Cancer cell ,Cancer research ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Plasmids ,Signal Transduction - Abstract
The serine/threonine kinases Akt1/PKBalpha, Akt2/PKBbeta, and Akt3/PKBgamma have been implicated in preventing cells from undergoing apoptosis. Although several small molecule inhibitors of Akt have been reported to induce apoptosis in cancer cells, these inhibitors may have additional targets. In the current study, we used an Akt3 small interfering RNA (Akt3 siRNA) to analyze apoptosis induction in Akt1 and Akt2 double knock-out mouse embryonic fibroblast cells (MEF-Akt1,2-DKO). Our data indicated that Akt3 siRNA inhibited Akt3 protein expression in a dose-dependent manner. As a result, phosphorylation of Akt and its downstream targets, including FKHRL1 and GSK3alpha/beta, were reduced accordingly. The treatment also induced apoptosis in MEF-Akt1,2-DKO cells. However, apoptosis induction is significant only when more than 80% of Akt3 protein was depleted. Reintroducing Akt3 totally rescued Akt3-siRNA-induced apoptosis in MEF-Akt1,2-DKO cells. In addition, reintroducing Akt1 also inhibited apoptosis induced by Akt3 siRNA. Moreover, Akt3 siRNA potentiated different stress-induced apoptosis in MEF-Akt1,2-DKO cells at a lower dose when compared with what is required for apoptosis induction by itself. Our study suggests that only a small portion of Akt is active in wild-type MEF cells and a threshold of Akt inhibition is required to induce apoptosis by pure Akt inhibitors. In addition, our data indicate that cells under stress require more Akt for its survival.
- Published
- 2006
- Full Text
- View/download PDF
36. Discovery and SAR of oxindole–pyridine-based protein kinase B/Akt inhibitors for treating cancers
- Author
-
Saul H. Rosenberg, Xuesong Liu, Jianchun Gong, Jennifer J. Bouska, Shayna R. Magnone, Tilman Oltersdorf, Ran Guan, Eric F. Johnson, Ken Jarvis, Alexander R. Shoemaker, Viraj B. Gandhi, Ron De Jong, Vered Klinghofer, Yan Luo, Yan Shi, Chang Park, Qun Li, Vincent L. Giranda, Anatol Oleksijew, and Gui-Dong Zhu
- Subjects
Models, Molecular ,Indoles ,Pyridines ,Clinical Biochemistry ,Pharmaceutical Science ,AKT1 ,Antineoplastic Agents ,Crystallography, X-Ray ,Biochemistry ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,GSK-3 ,Cell Line, Tumor ,Neoplasms ,Drug Discovery ,Animals ,Oxindole ,Protein kinase A ,Protein Kinase Inhibitors ,Molecular Biology ,Protein kinase B ,Cell Proliferation ,Dose-Response Relationship, Drug ,Molecular Structure ,biology ,Kinase ,Organic Chemistry ,Stereoisomerism ,Xenograft Model Antitumor Assays ,Oxindoles ,chemistry ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,Signal transduction ,Proto-Oncogene Proteins c-akt - Abstract
We describe a series of potent and selective oxindole–pyridine-based protein kinase B/Akt inhibitors. The most potent compound 11n in this series demonstrated an IC50 of 0.17 nM against Akt1 and more than 100-fold selectivity over other Akt isozymes. The selectivity against other protein kinases was highly dependent on the C-3 substitutions at the oxindole scaffold, with unsubstituted 9e or 3-furan-2-ylmethylene (11n) more selective and 3-(1H-pyrrol-2-yl)methylene (11f) or 3-(1H-imidazol-2-yl)methylene (11k) less selective. In a mouse xenograft model, 9d, 11f, and 11n inhibited tumor growth but with accompanying toxicity.
- Published
- 2006
- Full Text
- View/download PDF
37. Isoquinoline–pyridine-based protein kinase B/Akt antagonists: SAR and in vivo antitumor activity
- Author
-
Xuesong Liu, Keith W. Woods, Shayna R. Magnone, Jianchun Gong, Saul H. Rosenberg, Vincent L. Giranda, Jennifer J. Bouska, Eric F. Johnson, Gui-Dong Zhu, Anatol Oleksijew, Alexander R. Shoemaker, Viraj B. Gandhi, Tilman Oltersdorf, Vincent S. Stoll, Vered Klinghofer, Yan Luo, Akiyo Claiborne, Ron De Jong, Sheela A. Thomas, Yan Shi, Ran Guan, and Qun Li
- Subjects
Pyridines ,Clinical Biochemistry ,Pharmaceutical Science ,Antineoplastic Agents ,Pharmacology ,Crystallography, X-Ray ,Biochemistry ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,In vivo ,GSK-3 ,Cell Line, Tumor ,Drug Discovery ,Animals ,Humans ,Isoquinoline ,Protein kinase A ,Protein Kinase Inhibitors ,Molecular Biology ,Protein kinase B ,Molecular Structure ,biology ,Chemistry ,Organic Chemistry ,Isoquinolines ,Xenograft Model Antitumor Assays ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Lead compound - 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 iv t1/2 = 0.3 h, po F = 0%). Chlorination at C-1 position of the isoquinoline improved its pharmacokinetic property in mice (iv t1/2 = 5.0 h, po 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.
- Published
- 2006
- Full Text
- View/download PDF
38. Synthesis and structure–activity relationship of 3,4′-bispyridinylethylenes: Discovery of a potent 3-isoquinolinylpyridine inhibitor of protein kinase B (PKB/Akt) for the treatment of cancer
- Author
-
Saul H. Rosenberg, Tongmei Li, Gui-Dong Zhu, Keith W. Woods, Jason N. Abrams, Jürgen Dinges, Eric F. Johnson, Tilman Oltersdorf, Clarissa G. Jakob, Qun Li, John E. Fisher, Xuesong Liu, Vincent S. Stoll, Garrick Packard, Ron Des Jong, Sheela A. Thomas, Vincent L. Giranda, Xiaohong Song, Yan Luo, Vered Klinghofer, Jianchun Gong, and Yan Shi
- Subjects
Models, Molecular ,Pyridines ,Clinical Biochemistry ,Pharmaceutical Science ,AKT1 ,Antineoplastic Agents ,Biochemistry ,Cell Line ,Structure-Activity Relationship ,X-Ray Diffraction ,Neoplasms ,Drug Discovery ,medicine ,Staurosporine ,Protein kinase A ,Protein Kinase Inhibitors ,Molecular Biology ,Protein kinase B ,CAMK ,Chemistry ,Kinase ,Organic Chemistry ,Hydrogen Bonding ,embryonic structures ,Molecular Medicine ,Phosphorylation ,Proto-Oncogene Proteins c-akt ,Tyrosine kinase ,medicine.drug - Abstract
Structure-based design and synthesis of the 3,4'-bispyridinylethylene series led to the discovery of 3-isoquinolinylpyridine 13a as a potent PKB/Akt inhibitor with an IC(50) of 1.3nM against Akt1. Compound 13a shows excellent selectivity against distinct families of kinases such as tyrosine kinases and CAMK, and displays poor to marginal selectivity against closely related kinases in the AGC and CMGC families. Moreover, 13a demonstrates potent cellular activity comparable to staurosporine, with IC(50) values of 0.42 and 0.59microM against MiaPaCa-2 and the Akt1 overexpressing FL5.12-Akt1, respectively. Inhibition of phosphorylation of the Akt downstream target GSK3 was also observed in FL5.12-Akt1 cells with an EC(50) of 1.5microM. The X-ray structures of 12 and 13a in complex with PKA in the ATP-binding site were determined.
- Published
- 2006
- Full Text
- View/download PDF
39. Inhibition of the phosphatidylinositol 3-kinase/Akt pathway sensitizes MDA-MB468 human breast cancer cells to cerulenin-induced apoptosis
- Author
-
Yan Luo, Xuesong Liu, Yan Shi, and Vincent L. Giranda
- Subjects
Cancer Research ,Morpholines ,bcl-X Protein ,Down-Regulation ,Antineoplastic Agents ,Apoptosis ,Breast Neoplasms ,Inhibitor of apoptosis ,Amino Acid Chloromethyl Ketones ,Inhibitor of Apoptosis Proteins ,chemistry.chemical_compound ,Cytosol ,Humans ,Enzyme Inhibitors ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Inhibitor of apoptosis domain ,Caspase 3 ,Akt/PKB signaling pathway ,Chemistry ,Cytochromes c ,Caspase Inhibitors ,Cerulenin ,Mitochondria ,XIAP ,Enzyme Activation ,Oncogene Protein v-akt ,Proto-Oncogene Proteins c-bcl-2 ,Oncology ,Chromones ,Caspases ,Cancer research ,lipids (amino acids, peptides, and proteins) - Abstract
Fatty acid synthase is overexpressed in cancer especially in tumors with a poor prognosis. The specific fatty acid synthase inhibitor cerulenin can induce apoptosis in cancer cells. Likewise, phosphatidylinositol 3-kinase (PI3K)/Akt kinase activities are elevated in primary tumors and cancer cell lines. Here, we tested whether inhibition of PI3K/Akt pathway would sensitize cancer cells to cerulenin-induced apoptosis. We show that LY294002, an inhibitor of PI3K, sensitized MDA-MB468 breast cancer cells to cerulenin-induced apoptosis. In MDA-MB468 cells, cerulenin- and LY294002-mediated apoptosis was associated with caspase-3 activation and the release of cytochrome c from mitochondria to cytosol. In addition, we observed additional species of Bak in mitochondria, suggesting a possible Bak activation. Treatment of cells with cerulenin and LY294002 down-regulated the protein levels of X chromosome-linked inhibitor of apoptosis (XIAP), cellular inhibitor of apoptosis 1 (cIAP-1), and Akt, whereas the levels of mitogen-activated protein/extracellular signal-regulated kinase kinase and other antiapoptotic Bcl-2 family proteins (Bcl-2 and Bcl-xl) did not change. Interestingly, the nonspecific caspase inhibitor, z-VAD-FMK, inhibited the down-regulation of Akt, XIAP, and cIAP-1 in cerulenin- and LY294002-treated cells. In conclusion, these studies show that inhibition of PI3K can sensitize cerulenin-induced apoptosis in MBA-MB468 breast cancer cells via activation of caspases, down-regulation of antiapoptotic proteins, such as XIAP, cIAP-1 and Akt, and possibly, activation of Bak in mitochondria. [Mol Cancer Ther 2006;5(3):494–501]
- Published
- 2006
- Full Text
- View/download PDF
40. Optimal Classes of Chemotherapeutic Agents Sensitized by Specific Small-Molecule Inhibitors of Akt In Vitro and In Vivo
- Author
-
Thomas McGonigal, Keith W. Woods, Alexander R. Shoemaker, E. K.-H. Han, Yan Luo, Vered Klinghofer, Vincent L. Giranda, Anatol Oleksijew, Loren M. Lasko, Ran Guan, Xuesong Liu, John P. Fisher, Qun Li, Yan Shi, and Saul H. Rosenberg
- Subjects
Cancer Research ,Indazoles ,Indoles ,Paclitaxel ,medicine.drug_class ,Cell Survival ,synergy ,lcsh:RC254-282 ,Cell Line, Tumor ,inhibitors ,medicine ,PTEN ,Humans ,Doxorubicin ,Enzyme Inhibitors ,Protein kinase B ,PI3K/AKT/mTOR pathway ,biology ,Akt ,apoptosis ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,chemosensitization ,Kinetics ,Caspases ,Drug Design ,Cancer cell ,Cancer research ,biology.protein ,Phosphorylation ,Proto-Oncogene Proteins c-akt ,Camptothecin ,Topoisomerase inhibitor ,medicine.drug ,Research Article - Abstract
Akt is a serine/threonine kinase that transduces survival signals from survival/growth factors. Deregulation and signal imbalance in cancer cells make them prone to apoptosis. Upregulation or activation of Akt to aid the survival of cancer cells is a common theme in human malignancies. We have developed small-molecule Akt inhibitors that are potent and specific. These Akt inhibitors can inhibit Akt activity and block phosphorylation by Akt on multiple downstream targets in cells. Synergy in apoptosis induction was observed when Akt inhibitors were combined with doxorubicin or camptothecin. Akt inhibitor-induced enhancement of topoisomerase inhibitor cytotoxicity was also evident in long-term cell survival assay. Synergy with paclitaxel in apoptosis induction was evident in cells pretreated with paclitaxel, and enhancement of tumor delay by paclitaxel was demonstrated through cotreatment with Akt inhibitor Compound A (A-443654). Combination with other classes of chemotherapeutic agents did not yield any enhancement of cytotoxicity. These findings provide important guidance in selecting appropriate classes of chemotherapeutic agents for combination with Akt inhibitors in cancer treatment.
- Published
- 2005
41. Potent and selective inhibitors of Akt kinases slow the progress of tumors in vivo
- Author
-
Amanda K Mika, Eric F. Johnson, Joel D. Leverson, Keith W. Woods, Michael J. Mitten, Jennifer J. Bouska, Xuesong Liu, Bradley A. Zinker, Thomas McGonigal, Richard A. Smith, Saul H. Rosenberg, Tongmei Li, Mulugeta Mamo, Anatol Oleksijew, Ron De Jong, Tilman Oltersdorf, Vincent S. Stoll, Emily E. Gramling-Evans, Alexander R. Shoemaker, Yan Luo, Vered Klinghofer, Phong T. Nguyen, Jessica A. Powlas, Sheela A. Thomas, Yan Shi, Qun Li, Ran Guan, Edward K. Han, and Vincent L. Giranda
- Subjects
Models, Molecular ,Cancer Research ,Indazoles ,Indoles ,Pyridines ,AKT1 ,Antineoplastic Agents ,Mice, SCID ,Protein Serine-Threonine Kinases ,Pharmacology ,Carbohydrate metabolism ,Biology ,Sensitivity and Specificity ,Substrate Specificity ,Mice ,chemistry.chemical_compound ,In vivo ,Cell Line, Tumor ,Neoplasms ,Proto-Oncogene Proteins ,Animals ,Humans ,Phosphorylation ,Protein Kinase Inhibitors ,Protein kinase B ,Kinase ,Protein Structure, Tertiary ,Oncology ,Paclitaxel ,chemistry ,Tumor progression ,Disease Progression ,Signal transduction ,Proto-Oncogene Proteins c-akt - Abstract
The Akt kinases are central nodes in signal transduction pathways that are important for cellular transformation and tumor progression. We report the development of a series of potent and selective indazole-pyridine based Akt inhibitors. These compounds, exemplified by A-443654 (Ki = 160 pmol/L versus Akt1), inhibit Akt-dependent signal transduction in cells and in vivo in a dose-responsive manner. In vivo, the Akt inhibitors slow the progression of tumors when used as monotherapy or in combination with paclitaxel or rapamycin. Tumor growth inhibition was observed during the dosing interval, and the tumors regrew when compound administration was ceased. The therapeutic window for these compounds is narrow. Efficacy is achieved at doses ∼2-fold lower than the maximally tolerated doses. Consistent with data from knockout animals, the Akt inhibitors induce an increase in insulin secretion. They also induce a reactive increase in Akt phosphorylation. Other toxicities observed, including malaise and weight loss, are consistent with abnormalities in glucose metabolism. These data show that direct Akt inhibition may be useful in cancer therapy, but significant metabolic toxicities are likely dose limiting.
- Published
- 2005
- Full Text
- View/download PDF
42. Identification of Novel Binding Interactions in the Development of Potent, Selective 2-Naphthamidine Inhibitors of Urokinase. Synthesis, Structural Analysis, and SAR of N-Phenyl Amide 6-Substitution
- Author
-
Xumiao Zhao, Kent D. Stewart, Moshe Weitzberg, Michael D. Wendt, Vincent L. Giranda, Todd W. Rockway, Robert A. Mantei, Geyer Andrew George, Vicki L. Nienaber, Vered Klinghofer, and Mcclellan William J
- Subjects
Models, Molecular ,Stereochemistry ,medicine.drug_class ,Amidines ,Substituent ,Carboxamide ,Naphthalenes ,Crystallography, X-Ray ,Amidine ,Structure-Activity Relationship ,chemistry.chemical_compound ,Amide ,Drug Discovery ,medicine ,Humans ,Structure–activity relationship ,Phenyl group ,Binding site ,Binding Sites ,Molecular Structure ,Urokinase-Type Plasminogen Activator ,chemistry ,Solvents ,Molecular Medicine ,Salt bridge ,Protein Binding - Abstract
The preparation and assessment of biological activity of 6-substituted 2-naphthamidine inhibitors of the serine protease urokinase plasminogen activator (uPA, or urokinase) is described. 2-Naphthamidine was chosen as a starting point based on synthetic considerations and on modeling of substituent vectors. Phenyl amides at the 6-position were found to improve binding; replacement of the amide with other two-atom linkers proved ineffective. The phenyl group itself is situated near the S1' subsite; substitutions off of the phenyl group accessed S1' and other distant binding regions. Three new points of interaction were defined and explored through ring substitution. A solvent-exposed salt bridge with the Asp60A carboxylate was formed using a 4-alkylamino group, improving affinity to K(i) = 40 nM. Inhibitors also accessed two hydrophobic regions. One interaction is characterized by a tight hydrophobic fit made with a small dimple largely defined by His57 and His99; a weaker, less specific interaction involves alkyl groups reaching into the broad prime-side protein binding region near Val41 and the Cys42-Cys58 disulfide, displacing water molecules and leading to small gains in activity. Many inhibitors accessed two of these three regions. Affinities range as low as K(i) = 6 nM, and many compounds had K(i)100 nM, while moderate to excellent selectivity was gained versus four of five members of a panel of relevant serine proteases. Also, some selectivity against trypsin was generated via the interaction with Asp60A. X-ray structures of many of these compounds were used to inform our inhibitor design and to increase our understanding of key interactions. In combination with our exploration of 8-substitution patterns, we have identified a number of novel binding interactions for uPA inhibitors.
- Published
- 2003
- Full Text
- View/download PDF
43. Influenza Neuraminidase Inhibitors: Structure-Based Design of a Novel Inhibitor Series
- Author
-
Thomas J. Sowin, Minghua Sun, April Kennedy, Steven W. Muchmore, Gary Wang, Vincent S. Stoll, Yu-Gui Y. Gu, Clarence J. Maring, Chen Zhao, Graeme Laver, Kent D. Stewart, Dale J. Kempf, Darold L. Madigan, Jonathan Greer, Yuanwei Chen, Vincent L. Giranda, Hing L. Sham, Warren M. Kati, Ayda Saldivar, and Yibo Xu
- Subjects
Models, Molecular ,Pyrrolidines ,Molecular model ,Stereochemistry ,Neuraminidase ,Cyclopentanes ,Crystal structure ,Crystallography, X-Ray ,Ring (chemistry) ,Biochemistry ,Birds ,Structure-Activity Relationship ,chemistry.chemical_compound ,Zanamivir ,medicine ,Animals ,Combinatorial Chemistry Techniques ,Nanotechnology ,Amines ,Enzyme Inhibitors ,Binding Sites ,biology ,Active site ,Esters ,Stereoisomerism ,chemistry ,Influenza A virus ,Drug Design ,biology.protein ,Crystallization ,Protein crystallization ,Lead compound ,medicine.drug - Abstract
Combinatorial and structure-based medicinal chemistry strategies were used together to advance a lead compound with an activity of K(i) = 58 microM via a potency enhancement of70 000-fold to an analogue with an activity of K(i) = 0.8 nM against influenza neuraminidase (A/Tokyo/67). Lead optimization was initiated using molecular modeling and combinatorial chemistry. Protein crystal structures revealed that inconsistent structure-activity relationship (SAR) data resulted from different binding orientations of the inhibitor core five-membered rings from one series to another. Binding modes for a series of compounds showed up to a 180 degrees variation in orientation of the five-membered ring within the active site. Potent analogues were only achieved with chemical series that were observed to bind in the same orientation and yielded consistent SAR. In one series, consistent binding was obtained by an unprecedented occupation of a negatively charged binding pocket by a neutral methyl ester unit. The structural rationale for this novel SAR variation, based on protein crystallographic data, is given.
- Published
- 2003
- Full Text
- View/download PDF
44. Inhibitors of the Protease Domain of Urokinase-Type Plasminogen Activator
- Author
-
Vincent L. Giranda, Vicki L. Nienaber, and Todd W. Rockway
- Subjects
Models, Molecular ,Pharmacology ,Serine protease ,Urokinase ,Proteases ,Protease ,biology ,Chemistry ,Plasmin ,medicine.medical_treatment ,Protein degradation ,Urokinase-Type Plasminogen Activator ,Protein Structure, Tertiary ,Urokinase receptor ,Biochemistry ,Drug Discovery ,biology.protein ,medicine ,Animals ,Humans ,Protease Inhibitors ,Plasminogen activator ,medicine.drug - Abstract
Human urokinase-type plasminogen activator (uPA or uPA) has been implicated in the regulation and control of basement membrane and interstitial protein degradation. Since Urokinase plays a role in tissue remodeling, it may be responsible, in part, for the disease progression of cancer. Inhibitors of urokinase may then be useful in the treatment of cancer by retarding tumor growth and metastasis. Urokinase is a multidomain protein, two regions of the protein are most responsible for the observed proteolytic activity in cancer disease and progression. The N-terminal domain or ATF binds to a Urokinase receptor (uPAR) on the cell surface and the C-terminal serine protease domain, then, activates plasminogen to plasmin, beginning a cascade of events leading to the progression of cancer. Investigations of urokinase inhibition has been an area of ongoing research for the past 3 decades. It began with the discovery of small natural and unnatural amino acid derivatives or peptide analogs which exhibited weak inhibition of uPA. The last decade has seen the generation of several classes of potent and selective Urokinase inhibitor directed to the serine protease domain of the protein which have shown potential anti-cancer effects. The availability of structural information of enzyme-inhibitor complexes either by nuclear magnetic spectroscopy (NMR) or crystallography has allowed a detailed analysis of inhibitor protein interactions that contribute to observed inhibitor potency. Structural studies of specific inhibitor-uPA complexes will be discussed as well as the contributions of specific inhibitor protein interactions that are important for overall inhibitor potency. These data were used to discover a class of urokinase inhibitor based on the 2-Naphthamidine template that exhibits potent urokinase inhibition and excellent selectivity for urokinase over similar trypsin family serine proteases.
- Published
- 2002
- Full Text
- View/download PDF
45. Phase 3 study evaluating efficacy and safety of veliparib (V) plus carboplatin (Cb) or Cb in combination with standard neoadjuvant chemotherapy (NAC) in patients (pts) with early stage triple-negative breast cancer (TNBC)
- Author
-
Jens Huober, David Maag, Sibylle Loibl, Hope S. Rugo, William Fraser Symmans, Mehra Golshan, Danielle Sullivan, Gunter von Minckwitz, Charles E. Geyer, Michael Untch, Jose Juan Ponce Lorenzo, Kristi McIntyre, William M. Sikov, Xuan Liu, Priya Rastogi, Norman Wolmark, Mark D. McKee, Joyce O'Shaughnessy, Vincent L. Giranda, and Otto Metzger Filho
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Veliparib ,Cyclophosphamide ,medicine.medical_treatment ,Phases of clinical research ,Pharmacology ,Placebo ,Gastroenterology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Medicine ,Doxorubicin ,Triple-negative breast cancer ,Chemotherapy ,business.industry ,Carboplatin ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,business ,medicine.drug - Abstract
520 Background: Clinical studies suggest that TNBC is sensitive to DNA-damaging agents, including Cb. V is a potent PARP inhibitor that may enhance the antitumor activity of such agents. We present primary response data from a phase 3 randomized, placebo-controlled study (NCT02032277) evaluating the addition of V + Cb or Cb to neoadjuvant paclitaxel (P) followed by doxorubicin + cyclophosphamide (AC). Methods: Pts with histologically confirmed, invasive TNBC (T2–T4 N0–2 or T1 N1–2) amenable to surgical resection were randomized 2:1:1 to (Arm A) P 80 mg/m2 weekly + Cb AUC 6 mg/mL/min q3 weeks + V 50 mg PO BID; (Arm B) P + Cb + PO placebo; or (Arm C) P + IV placebo + PO placebo, for 12 weeks followed by AC (60 mg/m2 or 600 mg/m2 q2 or 3 weeks) × 4. Primary endpoint was pathologic complete response (pCR) in breast and nodes with > 80% power at 2-sided α of 0.05 using pair-wise comparisons for A vs B and A vs C to detect significant treatment effects using Χ2 test; secondary endpoint was rate of conversion to eligibility for breast conservation surgery (BCS). Adverse events (AEs) were assessed with NCI CTCAE V4.0. Results: Six hundred thirty-four pts (median age 50 years; range 22–79) were randomized to Arms A (n = 316), B (n = 160), or C (n = 158). Baseline characteristics were well balanced. No pCR difference was observed between Arms A and B (53.2% vs 57.5% p = 0.36), but pCR in Arm A was higher than Arm C (53.2% vs 31.0% p < 0.001). In non-prespecified analysis, pCR in Arm B was also higher than Arm C (57.5% vs 31.0% p < 0.001). Among pts ineligible for BCS at screening (n = 141), 62% were eligible after NAC in Arm A vs 44% each in Arms B (p = 0.13) and C (p = 0.14). Grade 3–4 AEs (Arms A/B/C, 86%/85%/45%) and serious AEs (30%/27%/14%) neutropenia, thrombocytopenia, anemia, nausea, and vomiting were increased with the addition of Cb; V did not impact toxicity. Median cycles of NAC were not reduced with V + Cb + P or Cb + P vs P. Conclusions: Addition of V to neoadjuvant Cb + P followed by AC did not increase pCR rate in breast and nodes in stage II–III TNBC, while addition of V + Cb or Cb alone to P followed by AC did. Cb (+/– V) increased toxicity but did not impact delivery of NAC. Clinical trial information: NCT02032277.
- Published
- 2017
- Full Text
- View/download PDF
46. Breast conservation after neoadjuvant chemotherapy for triple-negative breast cancer: Surgical results from an international randomized trial (BrighTNess)
- Author
-
Danielle Sullivan, Joyce O'Shaughnessy, Jens Huober, Mark D. McKee, William M. Sikov, Vincent L. Giranda, Hope S. Rugo, Charles E. Geyer, Mehra Golshan, David Maag, Michael Untch, Sibylle Loibl, Xuan Liu, Gunter von Minckwitz, and Norman Wolmark
- Subjects
Oncology ,Surgical results ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,Breast conservation ,business.industry ,medicine.medical_treatment ,medicine.disease ,Systemic therapy ,law.invention ,Breast cancer ,Randomized controlled trial ,law ,Internal medicine ,medicine ,Stage (cooking) ,business ,Triple-negative breast cancer - Abstract
514 Background: Neoadjuvant systemic therapy (NST) increases the frequency of breast-conserving therapy (BCT) in stage II-III breast cancer, but there is little data on how often it converts patients (pts) from BCT-ineligible (BCT-I) to BCT-eligible (BCT-E) and on the impact of other factors on surgical choices. We collected surgical assessment and management data from an international randomized trial of NST in triple-negative breast cancer (TNBC). Methods: Women with operable TNBC were randomized to veliparib (V) with carboplatin (C) and paclitaxel (P), placebo with C and P or placebo with P followed by doxorubicin and cyclophosphamide. The surgeons assessed BCT candidacy by clinico-radiographic criteria before and after NST; surgical management was at surgeon and patient discretion. We assessed interactions between BCT eligibility pre- and post-NST, germline BRCA mutation ( gBRCA) status, continent of treatment and achievement of pathologic complete response(pCR) and percentage of pts who underwent BCT versus mastectomy. Results: Pre- and post-NST surgical assessments were available for 604 pts who underwent surgery. BCT rates are listed in the Table. The BCT rate was 68% among pts deemed BCT-E after NST. pCR rates were identical between BCT-E pts who chose BCT (55%) vs. mastectomy (53%). Of 141 pts deemed BCT-I at baseline, 75 (53%) converted to BCT-E but only 42 (56%) of these opted for BCT. pCR rates were 49% in BCT-E converts vs. 36% in those remained BCT-I. gBRCA pts (n = 84) were less likely to choose BCT even if they were BCT-E. Pts treated in North America (NA) were less likely to choose BCT (55% vs. 80% for Europe and Asia P
- Published
- 2017
- Full Text
- View/download PDF
47. A phase I trial of veliparib (ABT-888) and temozolomide in children with recurrent CNS tumors: a pediatric brain tumor consortium report
- Author
-
Adekunle M. Adesina, Patrick A. Thompson, James M. Boyett, Alice P. Chen, Katherine E. Warren, Lindsay Kilburn, Xiao-Nan Li, Arzu Onar-Thomas, Mehmet Kocak, Evelyn McKeegan, Susan M. Blaney, Maryam Fouladi, Larry E. Kun, Jack Su, Brenda Chyla, Ian F. Pollack, Vincent L. Giranda, and Stewart Goldman
- Subjects
Adult ,Male ,Cancer Research ,Pediatric Brain Tumor Consortium ,Veliparib ,Adolescent ,Maximum Tolerated Dose ,medicine.medical_treatment ,Dacarbazine ,Biology ,Poly(ADP-ribose) Polymerase Inhibitors ,Poly (ADP-Ribose) Polymerase Inhibitor ,chemistry.chemical_compound ,Young Adult ,medicine ,Temozolomide ,Humans ,Child ,Antineoplastic Agents, Alkylating ,Medulloblastoma ,Chemotherapy ,Brain Neoplasms ,Infant ,medicine.disease ,Oncology ,chemistry ,Child, Preschool ,PARP inhibitor ,Immunology ,Cancer research ,Leukocytes, Mononuclear ,Benzimidazoles ,Female ,Neurology (clinical) ,Poly(ADP-ribose) Polymerases ,Pediatric Neuro-Oncology ,medicine.drug - Abstract
Most cytotoxic chemotherapy drugs induce single-strand DNA breaks, and radiation treatment induces double-strand DNA breaks. Repair of single-strand DNA breaks is mediated by multiple mechanisms, including the base-excision repair pathway, while double-strand DNA breaks are repaired by the homologous recombination (HR) and nonhomologous end-joining (NHEJ) repair pathways.1,2 Poly(ADP-ribose) polymerase (PARP) recognizes single- and double-strand DNA breaks3 and recruits and activates repair proteins from the base-excision,4 NHEJ,5,6 and HR repair pathways.4,7 Elevated PARP expression and/or activity may therefore mediate chemotherapy and radiation resistance and have indeed been demonstrated in numerous human cancers, including pediatric brain tumors.8–11 Veliparib (ABT-888) is an oral PARP inhibitor that potentiates anticancer activity of chemotherapy drugs,12,13 including temozolomide (TMZ), and enhances radiation efficacy.14–17 Several adult trials of veliparib and various chemotherapy agents,18–20 including TMZ,21,22 are ongoing. To explore the potential of veliparib as a novel treatment strategy in pediatric brain tumors, we previously showed that in a nonhuman primate model, the CSF-to-plasma ratio of veliparib was 57% ± 7%.23 We also demonstrated by immunohistochemistry (IHC) that PARP is highly expressed in pediatric medulloblastoma (MB) and glioblastoma multiforme (GBM) tumors but absent in postnatal normal brains, and veliparib treatment potently inhibited PARP activity in mouse orthotopic xenografts of pediatric MB and GBM and enhanced TMZ efficacy (X-N.L. and J.M.S., unpublished data). TMZ treatment in children with recurrent brain tumors showed only modest activity,24–26 with objective response rates ranging 5%–14%; although prior clinical data suggested a high rate of TMZ resistance in pediatric brain tumors, we hypothesized that such resistance to TMZ may be partially mediated by elevated PARP activity, and veliparib treatment may therefore abrogate PARP activity and improve response to TMZ. Here we report the results of our phase I trial of veliparib and TMZ in children with recurrent brain tumors. The primary objectives of the trial were: (i) to estimate the maximum tolerated dose (MTD) of veliparib and TMZ in children with recurrent brain tumors; (ii) to study veliparib pharmacokinetic (PK) and PARP inhibition in peripheral blood mononuclear cells (PBMCs); and (iii) to describe the toxicities of this regimen in children. The secondary objectives were: (i) to study PARP and other DNA repair protein expression by IHC in formalin-fixed, paraffin-embedded (FFPE) tumors; and (ii) to document tumor response to this regimen.
- Published
- 2014
48. Species Specificity of Amidine-Based Urokinase Inhibitors
- Author
-
Richard S. Smith, Xumiao Zhao, Aparna V. Sarthy, Vicki L. Nienaber, Kent D. Stewart, Moshe Weitzberg, K. I. Hulkower, Christopher C Butler, Vered Klinghofer, Vincent L. Giranda, Todd W. Rockway, Paul G. Richardson, Sarah A. Dorwin, Michael D. Wendt, and Tom Mcgonigal
- Subjects
Serine Proteinase Inhibitors ,Molecular Sequence Data ,Amidines ,Antineoplastic Agents ,Thiophenes ,Naphthalenes ,Crystallography, X-Ray ,Biochemistry ,Amiloride ,Carcinoma, Lewis Lung ,Mice ,Species Specificity ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Potency ,Amino Acid Sequence ,Binding site ,chemistry.chemical_classification ,Serine protease ,Urokinase ,Binding Sites ,Sequence Homology, Amino Acid ,biology ,Blood Proteins ,Urokinase-Type Plasminogen Activator ,Blood proteins ,Amino acid ,Enzyme ,chemistry ,biology.protein ,Sequence Alignment ,medicine.drug - Abstract
Inhibition of the proteolytic activity of urokinase has been shown to inhibit the progression of tumors in rodent models and is being investigated for use in human disease. Understanding the rodent/human species-specificity of urokinase inhibitors is therefore critical for interpretation of rodent cancer progression models that use these inhibitors. We report here studies with a panel of 11 diverse urokinase inhibitors in both human and mouse enzymatic assays. Inhibitors such as amiloride, B428, and naphthamidine, that occupy only the S1 subsite pocket were found to be nearly equipotent between the human and the murine enzymes. Inhibitors that access additional, more distal, pockets were significantly more potent against the human enzyme but there was no corresponding potency increase against the murine enzyme. X-ray crystallographic structures of these compounds bound to the serine protease domain of human urokinase were solved and examined in order to explain the human/mouse potency differences. The differences in inhibitor potency could be attributed to four amino acid residues that differ between murine and human urokinases: 60, 99, 146, and 192. These residues are Asp, His, Ser, and Gln in human and Gln, Tyr, Glu, and Lys in mouse, respectively. Compounds bearing a cationic group that interacts with residue 60 will preferentially bind to the human enzyme because of favorable electrostatic interactions. The hydrogen bonding to residue 192 and steric considerations with residues 99 and 146 also contribute to the species specificity. The nonparallel human/mouse enzyme inhibition observations were extended to a cell-culture assay of urokinase-activated plasminogen-mediated fibronectin degradation with analogous results. These studies will aid the interpretation of in vivo evaluation of urokinase inhibitors.
- Published
- 2001
- Full Text
- View/download PDF
49. Downregulation of Akt1 Inhibits Anchorage-Independent Cell Growth and Induces Apoptosis in Cancer Cells
- Author
-
Yan Luo, Yan Shi, Vincent L. Giranda, Shi-Chung Ng, Zehan Chen, Edward K. Han, Saul H. Rosenberg, and Xuesong Liu
- Subjects
oligonucleotide ,Cancer Research ,Cell Survival ,antisense ,Blotting, Western ,AKT1 ,Down-Regulation ,AKT2 ,Antineoplastic Agents ,Cytochrome c Group ,Biology ,Protein Serine-Threonine Kinases ,lcsh:RC254-282 ,AKT3 ,Colony-Forming Units Assay ,Cancer stem cell ,Proto-Oncogene Proteins ,Oxazines ,medicine ,Cell Adhesion ,Tumor Cells, Cultured ,Humans ,cancer ,Coloring Agents ,Protein kinase B ,Akt1 ,Cell growth ,apoptosis ,Cancer ,Fibroblasts ,Oligonucleotides, Antisense ,medicine.disease ,Flow Cytometry ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Xanthenes ,Caspases ,Cancer cell ,embryonic structures ,Cancer research ,combination treatment ,Poly(ADP-ribose) Polymerases ,Proto-Oncogene Proteins c-akt ,Cell Division ,Research Article - Abstract
The serine/threonine kinases, Akt1/PKBalpha, Akt2/PKBbeta, and Akt3/PKBgamma, play a critical role in preventing cancer cells from undergoing apoptosis. However, the function of individual Akt isoforms in the tumorigenicity of cancer cells is still not well defined. In the current study, we used an Akt1 antisense oligonucleotide (AS) to specifically downregulate Akt1 protein in both cancer and normal cells. Our data indicate that Akt1 AS treatment inhibits the ability of MiaPaCa-2, H460, HCT-15, and HT1080 cells to grow in soft agar. The treatment also induces apoptosis in these cancer cells as demonstrated by FACS analysis and a caspase activity assay. Conversely, Akt1 AS treatment has little effect on the cell growth and survival of normal human cells including normal human fibroblast (NHF), fibroblast from muscle (FBM), and mammary gland epithelial 184B5 cells. In addition, Akt1 AS specifically sensitizes cancer cells to typical chemotherapeutic agents. Thus, Akt1 is indispensable for maintaining the tumorigenicity of cancer cells. Inhibition of Akt1 may provide a powerful sensitization agent for chemotherapy specifically in cancer cells.
- Published
- 2001
50. Discovering novel ligands for macromolecules using X-ray crystallographic screening
- Author
-
Paul L. Richardson, Vicki L. Nienaber, Vincent L. Giranda, Vered Klighofer, Jennifer J. Bouska, and Jonathan Greer
- Subjects
Time Factors ,Macromolecular Substances ,Computer science ,Fragment-based lead discovery ,Drug Evaluation, Preclinical ,Molecular Conformation ,Biomedical Engineering ,Administration, Oral ,Antineoplastic Agents ,Bioengineering ,Computational biology ,Naphthalenes ,Crystallography, X-Ray ,Ligands ,Proteomics ,Applied Microbiology and Biotechnology ,Structure-Activity Relationship ,chemistry.chemical_compound ,Lead (geology) ,Pharmaceutical technology ,Enzyme Inhibitors ,Hit to lead ,Urokinase-Type Plasminogen Activator ,Identification (information) ,chemistry ,Drug Design ,Quinolines ,Molecular Medicine ,Lead compound ,Biotechnology ,Macromolecule - Abstract
The need to decrease the time scale for clinical compound discovery has led to innovations at several stages in the process, including genomics/proteomics for target identification, ultrahigh-throughput screening for lead identification, and structure-based drug design and combinatorial chemistry for lead optimization. A critical juncture in the process is the identification of a proper lead compound, because a poor choice may generate costly difficulties at later stages. Lead compounds are commonly identified from high-throughput screens of large compound libraries, derived from known substrates/inhibitors, or identified in computational prescreeusing X-ray crystal structures. Structural information is often consulted to efficiently optimize leads, but under the current paradigm, such data require preidentification and confirmation of compound binding. Here, we describe a new X-ray crystallography-driven screening technique that combines the steps of lead identification, structural assessment, and optimization. The method is rapid, efficient, and high-throughput, and it results in detailed crystallographic structure information. The utility of the method is demonstrated in the discovery and optimization of a new orally available class of urokinase inhibitors for the treatment of cancer.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.