24 results on '"Stanley C. Gill"'
Search Results
2. Supplementary Figure Legend from Effect of Intermittent Dosing Regimens of Erlotinib on Methylnitrosourea-Induced Mammary Carcinogenesis
- Author
-
Clinton J. Grubbs, Holly L. Nicastro, M. Margaret Juliana, Vernon E. Steele, Ann Bode, Chris Tucker, Stanley C. Gill, Kenneth K. Iwata, Eva Szabo, and Ronald A. Lubet
- Abstract
PDF file - 50K
- Published
- 2023
3. Phase 3 Trial of Gilteritinib Plus Azacitidine Vs Azacitidine for Newly Diagnosed FLT3mut+ AML Ineligible for Intensive Chemotherapy
- Author
-
Eunice S. Wang, Pau Montesinos, Mark D. Minden, Je-Hwan Lee, Michael Heuser, Tomoki Naoe, Wen-Chien Chou, Kamel Laribi, Jordi Esteve, Jessica K. Altman, Violaine Havelange, Anne-Marie Watson, Carlo Gambacorti-Passerini, Elzbieta Patkowska, Shufang Liu, Ruishan Wu, Nisha Philipose, Jason E. Hill, Stanley C. Gill, Elizabeth Shima Rich, Ramon V. Tiu, Wang, E, Montesinos, P, Minden, M, Lee, J, Heuser, M, Naoe, T, Chou, W, Laribi, K, Esteve, J, Altman, J, Havelange, V, Watson, A, Gambacorti-Passerini, C, Patkowska, E, Liu, S, Ruishan, W, Philipose, N, Hill, J, Gill, S, Rich, E, Tiu, R, and UCL - SSS/DDUV/MEXP - Médecine expérimentale
- Subjects
Adult ,Leukemia, Myeloid, Acute ,Pyrazines ,Antineoplastic Combined Chemotherapy Protocols ,Immunology ,Azacitidine ,Humans ,Clinical Trials, Acute Myeloid Leukemia, Azacitidine, Gilterinib, Intensive Chemotherapy ,Cell Biology ,Hematology ,Biochemistry ,Aged - Abstract
Treatment results for patients with newly diagnosed FMS-like tyrosine kinase 3 (FLT3)-mutated (FLT3mut+) acute myeloid leukemia (AML) ineligible for intensive chemotherapy are disappointing. This multicenter, open-label, phase 3 trial randomized (2:1) untreated adults with FLT3mut+ AML ineligible for intensive induction chemotherapy to receive gilteritinib (120 mg/d orally) and azacitidine (GIL + AZA) or azacitidine (AZA) alone. The primary end point was overall survival (OS). At the interim analysis (August 26, 2020), a total of 123 patients were randomized to treatment (GIL + AZA, n = 74; AZA, n = 49). Subsequent AML therapy, including FLT3 inhibitors, was received by 20.3% (GIL + AZA) and 44.9% (AZA) of patients. Median OS was 9.82 (GIL + AZA) and 8.87 (AZA) months (hazard ratio, 0.916; 95% CI, 0.529-1.585; P = .753). The study was closed based on the protocol-specified boundary for futility. Median event-free survival was 0.03 month in both arms. Event-free survival defined by using composite complete remission (CRc) was 4.53 months for GIL + AZA and 0.03 month for AZA (hazard ratio, 0.686; 95% CI, 0.433-1.087; P = .156). CRc rates were 58.1% (GIL + AZA) and 26.5% (AZA) (difference, 31.4%; 95% CI, 13.1-49.7; P < .001). Adverse event (AE) rates were similar for GIL + AZA (100%) and AZA (95.7%); grade ≥3 AEs were 95.9% and 89.4%, respectively. Common AEs with GIL + AZA included pyrexia (47.9%) and diarrhea (38.4%). Gilteritinib steady-state trough concentrations did not differ between GIL + AZA and gilteritinib. GIL + AZA resulted in significantly higher CRc rates, although similar OS compared with AZA. Results support the safety/tolerability and clinical activity of upfront therapy with GIL + AZA in older/unfit patients with FLT3mut+ AML. This trial was registered at www.clinicaltrials.gov as #NCT02752035.
- Published
- 2022
4. AML-256 A Phase 1 Study of Gilteritinib in Combination With Induction and Consolidation Chemotherapy in Patients With Newly Diagnosed Acute Myeloid Leukemia: Final Study Results
- Author
-
Keith W. Pratz, Mohamad Cherry, Nikolai A. Podoltsev, Jessica K. Altman, Alexander E. Perl, Brenda W. Cooper, Joseph G. Jurcic, Tara L. Lin, Gary J. Schiller, Ruishan Wu, Jason E. Hill, Stanley C. Gill, Angela James, Elizabeth Shima Rich, Nahla Hasabou, and Mark J. Levis
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
5. Abstract CT537: A phase 2, multicenter, randomized, double-blind trial of maintenance therapy with FLT3 inhibitor gilteritinib (ASP2215) in patients with FLT3/ITD AML (GOSSAMER study)
- Author
-
Mark D. Minden, Jacob M. Rowe, Emmanuel Gyan, Kohmei Kubo, Nahla Hasabou, David Delgado, Wensheng He, Stanley C. Gill, Jason E. Hill, and Ramon Tiu
- Subjects
Cancer Research ,Oncology - Abstract
Background: Patients with acute myeloid leukemia (AML) in remission are at high risk of relapse, warranting remission-prolonging therapy. Gilteritinib is the first FMS-like tyrosine kinase 3 (FLT3) inhibitor approved as monotherapy in FLT3-mutated relapsed/refractory AML. The aim of the study was to compare relapse-free survival (RFS) in patients with FLT3/internal tandem duplication AML in first complete remission who received gilteritinib or placebo. Method: In this phase 2, double-blind trial, patients were randomized 2:1 to gilteritinib (120 mg) or placebo once daily for up to 2 years, beginning after completion of induction/consolidation (I/C) therapy. RFS, defined as time from randomization date until the date of documented relapse or death from any cause was assessed via independent review committee adjudication (primary efficacy endpoint). Overall survival (OS) was defined as the time from date of randomization until date of death from any cause. Survival and treatment-emergent adverse events (TEAEs) were assessed 30 days after treatment discontinuation, and every 3 months thereafter. Due to a slow accrual rate, this trial (NCT02927262) was changed from a planned phase 3 to phase 2 based on the reduction in sample size. Results: 124 patients were screened and 98 were randomized (gilteritinib 63, placebo 35) and are included in the full analysis set (FAS), of whom 32 (gilteritinib 20, placebo 12) completed 2 years of treatment. The safety analysis set (SAF) comprised 97 patients (gilteritinib 62, placebo 35) who had received ≥1 dose of study drug. In the FAS, relapse occurred in 31/63 (49.2%) gilteritinib- and 20/35 (57.1%) placebo-treated patients; 3/63 (4.8%) gilteritinib-treated patients died without relapse. RFS was not significantly improved with gilteritinib vs placebo (hazard ratio [HR] 0.738, 95% confidence interval [CI] 0.407-1.336; 1-sided stratified log-rank p-value 0.163). Median RFS was 24.02 months for gilteritinib and 15.84 months for placebo. OS in the FAS was not significantly different between treatments (gilteritinib 21/63, [33.3%], placebo 11/35 [31.4%]; HR 1.130, 95% CI 0.540-2.364; 1-sided stratified log-rank p-value 0.627). Median OS was not reached in either arm. TEAEs in the SAF occurred in 58/62 (93.5%) gilteritinib- and 33/35 (94.3%) placebo-treated patients including, respectively, 51/62 (82.3%) and 20/35 (57.1%) drug-related TEAEs and 10/62 (16.1%) and 3/35 (8.6%) serious drug-related TEAEs. The most common TEAEs were increased blood creatine phosphokinase (gilteritinib 18/62 [29.0%], placebo 1/35 [2.9%]) and thrombocytopenia (gilteritinib 12/62 [19.4%], placebo 4/35 [11.4%]). Conclusion: Gilteritinib showed improved RFS versus placebo, but the difference was not statistically significant. There were no new safety findings in the gilteritinib arm. Citation Format: Mark D. Minden, Jacob M. Rowe, Emmanuel Gyan, Kohmei Kubo, Nahla Hasabou, David Delgado, Wensheng He, Stanley C. Gill, Jason E. Hill, Ramon Tiu. A phase 2, multicenter, randomized, double-blind trial of maintenance therapy with FLT3 inhibitor gilteritinib (ASP2215) in patients with FLT3/ITD AML (GOSSAMER study) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr CT537.
- Published
- 2022
6. Mass balance, pharmacokinetics, and metabolism of linsitinib in cancer patients
- Author
-
Jorge Chaves, Stanley C. Gill, Elizabeth Conklin, Srinivasu Poondru, Margaret Singh, Barbara Parker, Masanori Nagata, and Geoffrey Yuen
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Linsitinib ,Urine ,Pharmacology ,Toxicology ,Excretion ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacokinetics ,Neoplasms ,Humans ,Pharmacology (medical) ,Adverse effect ,Protein Kinase Inhibitors ,Feces ,Aged ,Chemistry ,Imidazoles ,Middle Aged ,030104 developmental biology ,Renal Elimination ,Oncology ,Tolerability ,030220 oncology & carcinogenesis ,Pyrazines ,Female ,Metabolic Networks and Pathways - Abstract
This study characterized the pharmacokinetics, mass balance, routes and extent of elimination, metabolites, and safety of a single oral dose of 14C-linsitinib, an IGF-1R/IR inhibitor, in patients with advanced solid tumors. The tolerability of linsitinib after multiple-dose administration was assessed in those patients who wished to continue treatment beyond the single 14C-linsitinib dose. Five patients received a single oral dose of 150 mg 14C-linsitinib, followed by collection of blood, plasma, urine, and feces for 10 days. The collected material was analyzed for total radioactivity, linsitinib, and metabolites. The safety of 150 mg of unlabeled linsitinib administered twice daily until disease progression was also assessed. The median time to reach the maximum plasma concentration of linsitinib was 3.0 h, median maximum plasma concentration was 1789 ng/mL, median terminal half-life was 2.4 h, and median apparent oral clearance was 12.45 L/h. After a single dose of 14C-linsitinib, 5.44 and 76.4 % of mean total radioactivity administered were recovered in urine and feces, respectively. Eighteen linsitinib metabolites (M1–M18) were detected in plasma, urine, and feces samples, and their structures were elucidated. The main metabolic reactions of linsitinib in humans were oxidation and sulfate conjugation. Linsitinib was well tolerated after a single dose of 14C-linsitinib, and fatigue was the most frequent adverse event following multiple doses of unlabeled linsitinib. 14C-linsitinib is rapidly absorbed and extensively metabolized. Linsitinib excretion via bile into feces is the predominant elimination route from plasma with minor renal elimination.
- Published
- 2016
7. Pharmacokinetic-Pharmacodynamic Assessment of Faropenem in a Lethal Murine Bacillus anthracis Inhalation Postexposure Prophylaxis Model
- Author
-
Nebojsa Janjic, Lynda L. Miller, Ian A. Critchley, Jinfang Li, Amber A. Beaudry, Christopher M. Rubino, Sujata M. Bhavnani, Paul G. Ambrose, Stanley C. Gill, Jennifer Bassett, Kimberly Clawson Stone, and Henry S. Heine
- Subjects
Pharmacology ,Biology ,beta-Lactams ,Models, Biological ,beta-Lactamases ,Anthrax ,Mice ,chemistry.chemical_compound ,Pharmacokinetics ,Animals ,Experimental Therapeutics ,Pharmacology (medical) ,Antibacterial agent ,Inhalation exposure ,Inhalation Exposure ,Mice, Inbred BALB C ,Dose-Response Relationship, Drug ,Inhalation ,Lethal dose ,Faropenem ,Blood Proteins ,Effective dose (pharmacology) ,Anti-Bacterial Agents ,Disease Models, Animal ,Infectious Diseases ,chemistry ,Bacillus anthracis ,Pharmacodynamics ,Female - Abstract
There are few options for prophylaxis after exposure to Bacillus anthracis , especially in children and women of childbearing potential. Faropenem is a β-lactam in the penem subclass that is being developed as an oral prodrug, faropenem medoxomil, for the treatment of respiratory tract infections. Faropenem was shown to have in vitro activity against B. anthracis strains that variably express the bla1 β-lactamase (MIC range, ≤0.06 to 1 μg/ml). In this study we evaluated the pharmacokinetic-pharmacodynamic (PK-PD) relationships between the plasma faropenem free-drug ( f ) concentrations and efficacy against B. anthracis in a murine postexposure prophylaxis inhalation model. The plasma PKs and PKs-PDs of faropenem were evaluated in BALB/c mice following the intraperitoneal (i.p.) administration of doses ranging from 2.5 to 160 mg/kg of body weight. For the evaluation of efficacy, mice received by inhalation aerosol doses of B. anthracis (Ames strain; faropenem MIC, 0.06 μg/ml) at 100 times the 50% lethal dose. The faropenem dosing regimens (10, 20, 40, and 80 mg/kg/day) were administered i.p. at 24 h postchallenge at 4-, 6-, and 12-h intervals for 14 days. The sigmoid maximum-threshold-of-efficacy ( E max ) model fit the survival data, in which the free-drug area under the concentration-time curve ( f AUC)/MIC ratio, the maximum concentration of free drug in plasma ( fC max )/MIC ratio, and the cumulative percentage of a 24-h period that the free-drug concentration exceeds the MIC under steady-state pharmacokinetic conditions ( f % T MIC ) were each evaluated. Assessment of f % T MIC demonstrated the strongest correlation with survival ( R 2 = 0.967) compared to the correlations achieved by assessment of f AUC/MIC or fC max /MIC, for which minimal correlations were observed. The 50% effective dose (ED 50 ), ED 90 , and ED 99 corresponded to f % T MIC values of 10.6, 13.4, and 16.4%, respectively, and E max was 89.3%. Overall, faropenem demonstrated a high level of activity against B. anthracis in the murine postexposure prophylaxis inhalation model.
- Published
- 2010
8. Final Results of the Chrysalis Trial: A First-in-Human Phase 1/2 Dose-Escalation, Dose-Expansion Study of Gilteritinib (ASP2215) in Patients with Relapsed/Refractory Acute Myeloid Leukemia (R/R AML)
- Author
-
Eunice S. Wang, Stuart L. Goldberg, Alexander I. Spira, Catherine C. Smith, Joseph G. Jurcic, Celalettin Ustun, Harry P. Erba, Mark R. Litzow, Andreas Neubauer, Giovanni Martinelli, Maria R. Baer, Raoul Tibes, Mark J. Levis, Erkut Bahceci, Charles Liu, Richard A. Larson, Stanley C. Gill, Jessica K. Altman, Ellen K. Ritchie, Robert K. Stuart, David F. Claxton, Jorge E. Cortes, Christoph Röllig, Stephen A. Strickland, Gary J. Schiller, and Alexander E. Perl
- Subjects
medicine.medical_specialty ,business.industry ,Immunology ,Gilteritinib ,Cell Biology ,Hematology ,PK Parameters ,First in human ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Tolerability ,030220 oncology & carcinogenesis ,Internal medicine ,Relapsed refractory ,Dose escalation ,medicine ,In patient ,business ,Bristol-Myers ,health care economics and organizations ,030215 immunology - Abstract
Background: Gilteritinib (ASP2215) is a novel, highly selective, potent oral FLT3/AXL inhibitor with preclinical activity against FLT3-ITD activating and FLT3-D835 resistance mutations. The objectives of this phase 1/2 study were to assess gilteritinib safety/tolerability, pharmacokinetic (PK) and pharmacodynamic (PD) profiles after single- and multiple-day dosing, and antileukemic effects in patients with R/R AML. Methods: This open-label study (NCT02014558) enrolled patients (≥18 yr) into 1 of 7 dose-escalation cohorts (20-450 mg once daily [QD]) or concomitant dose-expansion cohorts. While confirmed FLT3 mutation was not an inclusion criterion, each expanded dose level enrolled ≥10 patients with FLT3 mutations (FLT3mut+); 120 and 200 mg dose levels were further expanded with ≥40 FLT3mut+ patients. The choice to expand these dose cohorts was based upon FLT3 inhibition in correlative assays and clinical activity seen during dose escalation. Safety and tolerability were primary endpoints; blood samples were drawn from patients in the dose-escalation cohorts to evaluate gilteritinib PK parameters and PD effects. Antileukemic response rates (eg, complete remission [CR], CR with incomplete platelet recovery [CRp], CR with incomplete hematological recovery [CRi], overall response rate [ORR]) were secondary endpoints. Results: Patients (N=252; 129M:123F, median age 62 yr [range: 21-90]) enrolled between October 2013 and August 2015 received ≥1 dose of gilteritinib. The study population was heavily pretreated: 70% (n=177) had ≥2 prior AML therapies, 29% (n=73) had a prior stem cell transplant, and 25% (n=63) had prior TKI treatment with sorafenib most commonly used. Across the study, 194 patients had a locally confirmed FLT3 mutation (ITD, n=159; D835, n=13; ITD-D835, n=16; other, n=6). For all enrolled patients, progressive disease (n=75), lack of efficacy (n=44), adverse events (n=34), and death (n=29) were the most common reasons for treatment discontinuation. Seven deaths were considered possibly/probably related to treatment: pulmonary embolism, respiratory failure, hemoptysis, intracranial bleed, ventricular fibrillation, septic shock, and neutropenia (all n=1). Maximum tolerated dose was determined to be 300 mg when 2 of 3 patients in the 450 mg cohort experienced diarrhea and/or hepatic transaminase elevation as dose-limiting toxicities. Diarrhea (16%) and fatigue (15%) were the most commonly reported treatment-related adverse events of any grade. Less than 5% of patients (11/252) had a maximum post-baseline QTcF interval >500 msec. Gilteritinib concentrations were generally dose proportional and showed both a long-elimination half-life (45-159 h) and substantial accumulation (3.2-10 fold) by day 15. An exposure-related increase in the inhibition of FLT3 phosphorylation with increasing doses of gilteritinib was also observed. Gilteritinib showed strong antileukemic activity in FLT3mut+ patients (ORR=49%); response was observed less frequently in patients with wild-type FLT3 (ORR=12%). While CR, CRi, and CRp occurred at all doses, responses were enriched among FLT3mut+ patients with gilteritinib steady-state trough concentrations ≥100 ng/mL, which correlated with potent FLT3 inhibition in PD assays and corresponded to doses ≥80 mg. The ORR in 169 FLT3mut+ patients receiving ≥80 mg was 52% (Table); median overall survival in this patient population was ~31 wk (range: 1.7-61; Figure) and median duration of response was 20 wk (range: 1.1-55). Clinical responses occurred in FLT3mut+ patients with -ITD, -D835, and both mutations (ORR: 55%, 17%, and 62%, respectively) as well as in FLT3mut+ patients with or without prior TKI treatment (ORR: 42% vs 56%, respectively). Conclusions: This PD-driven, first-in-human study shows that gilteritinib was well tolerated and generated frequent, prolonged, clinically important responses in FLT3mut+ patients with R/R AML. Antileukemic responses were enriched in FLT3mut+ patients treated at doses that consistently and potently inhibited FLT3 phosphorylation. The survival of these patients appears better than expected for this patient population when treated with standard therapy. Our data suggest that FLT3 inhibition may improve survival in patients with FLT3mut+R/R AML; as such, phase 3 testing of oral gilteritinib 120 mg QD in patients with FLT3mut+R/R AML after first-line therapy is underway (NCT02421939). Disclosures Perl: Astellas US Pharma Inc.: Consultancy, Membership on an entity's Board of Directors or advisory committees; Daichi Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees; Asana Biosciences: Consultancy, Membership on an entity's Board of Directors or advisory committees; Arog Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Actinium Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees. Altman:Janssen: Other: advisory board; BMS: Membership on an entity's Board of Directors or advisory committees; Spectrum: Other: advisory board; Ariad: Other: advisory board; Seattle Genetics: Other: advisory board; Syros: Other: advisory board. Cortes:ARIAD: Consultancy, Research Funding; BMS: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding; Teva: Research Funding. Smith:Astellas: Research Funding. Erba:Jannsen: Consultancy, Research Funding; Millennium Pharmaceuticals, Inc.: Research Funding; Agios: Research Funding; Juno: Research Funding; Incyte: Consultancy, DSMB, Speakers Bureau; Daiichi Sankyo: Consultancy; Ariad: Consultancy; Amgen: Consultancy, Research Funding; Astellas: Research Funding; Gylcomimetics: Other: DSMB; Seattle Genetics: Consultancy, Research Funding; Sunesis: Consultancy; Novartis: Consultancy, Speakers Bureau; Celator: Research Funding; Celgene: Consultancy, Speakers Bureau; Pfizer: Consultancy. Gill:Astellas: Employment. Goldberg:Bristol Myers Squibb, Novartis: Speakers Bureau; Novartis: Consultancy; COTA Inc: Employment; Pfizer: Honoraria; Neostem: Equity Ownership. Jurcic:Astellas: Research Funding. Larson:Astellas: Consultancy, Research Funding; Bristol-Myers Squibb: Consultancy. Liu:Astellas: Employment. Ritchie:Celgene: Consultancy, Other: Travel, Accomodations, Expenses, Speakers Bureau; Incyte: Consultancy, Speakers Bureau; Novartis: Consultancy, Other: Travel, Accommodations, Expenses, Research Funding, Speakers Bureau; Ariad: Speakers Bureau; Pfizer: Consultancy, Research Funding; Astellas Pharma: Research Funding; Bristol-Meyers Squibb: Research Funding; NS Pharma: Research Funding. Schiller:Incyte Corporation: Research Funding. Strickland:Celator: Research Funding; Cyclacel: Research Funding; Karyopharm Therapeutica: Research Funding; GlaxoSmithKline: Research Funding; Baxalta: Consultancy; Boehringer Ingelheim: Consultancy, Research Funding; Ambit: Consultancy; Alexion Pharmaceuticals: Consultancy; Astellas Pharma: Research Funding; CTI Biopharma: Consultancy; Daiichi Sankyo: Consultancy; Sunesis Pharmaceuticals: Consultancy, Research Funding; Abbvie: Research Funding; Sanofi: Research Funding. Wang:Incyte: Speakers Bureau; Immunogen: Research Funding. Stuart:Sunesis: Consultancy, Honoraria, Other: Travel, Accomodations, Expenses, Research Funding; Agios: Research Funding; Incyte: Research Funding; Bayer: Research Funding; Celator: Research Funding; Astellas: Research Funding. Martinelli:Ariad: Consultancy, Speakers Bureau; Pfizer: Consultancy, Speakers Bureau; BMS: Speakers Bureau; Roche: Consultancy, Speakers Bureau; MSD: Consultancy; Genentech: Consultancy; Novartis: Speakers Bureau; Celgene: Consultancy, Speakers Bureau; Amgen: Consultancy, Speakers Bureau. Bahceci:Astellas: Employment. Levis:Millennium: Consultancy, Research Funding; Astellas: Consultancy, Honoraria, Research Funding; Daiichi-Sankyo: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Research Funding.
- Published
- 2016
9. [Untitled]
- Author
-
Joyce Nelson, Kerry Nixon, Stanley C. Gill, James A. Farmer, Mark B. Judkins, Philip M. Zack, Richard Bolin, Daniel W. Drolet, Chris Tucker, Julie Wolf, and Raymond A. Bendele
- Subjects
Pharmacology ,Anti vegf ,medicine.medical_specialty ,genetic structures ,business.industry ,Aptamer ,Organic Chemistry ,Pharmaceutical Science ,Absorption (skin) ,eye diseases ,Vascular endothelial growth factor A ,Endocrinology ,Pharmacokinetics ,Internal medicine ,Toxicity ,Blood plasma ,medicine ,Pegaptanib Sodium ,Molecular Medicine ,Pharmacology (medical) ,sense organs ,business ,Biotechnology - Abstract
Purpose. The objective of this study was to determine the pharmacokinetics and safety for NX1838 following injection into the vitreous humor of rhesus monkeys.
- Published
- 2000
10. Detection and plasma pharmacokinetics of an anti-vascular endothelial growth factor oligonucleotide-aptamer (NX1838) in rhesus monkeys
- Author
-
Mark B. Judkins, Stanley C. Gill, Long-Shiuh Chen, Chris Tucker, Daniel W. Drolet, and James A. Farmer
- Subjects
Quality Control ,Vascular Endothelial Growth Factor A ,Angiogenesis ,Injections, Subcutaneous ,Aptamer ,Oligonucleotides ,Endothelial Growth Factors ,Polyethylene glycol ,chemistry.chemical_compound ,Drug Stability ,Pharmacokinetics ,Blood plasma ,Animals ,Humans ,Chromatography, High Pressure Liquid ,Lymphokines ,Chromatography ,Vascular Endothelial Growth Factors ,Chemistry ,Reproducibility of Results ,General Chemistry ,Macaca mulatta ,In vitro ,Vascular endothelial growth factor ,Injections, Intravenous ,Quantitative analysis (chemistry) - Abstract
Aptamers are oligonucleotide ligands selected, in vitro, to bind a specified target protein. The first aptamer to reach human clinical testing is NX1838, a polyethylene glycol conjugated aptamer that inhibits vascular endothelial growth factor. This paper describes the validation of a high-performance liquid chromatographic anion-exchange method for the determination of NX1838 in plasma. Measurements of intact NX1838 had a coefficient of variation of less than 8% and an accuracy between 107% and 115%. The assay was utilized to determine NX1838 plasma pharmacokinetics in rhesus monkeys following a single 1 mg/kg intravenous or subcutaneous dose. Following intravenous administration, the maximum achieved plasma concentration was 25.5 μg/ml with a terminal half-life of 9.3 h and clearance rate of 6.2 ml/h. After subcutaneous administration, the fraction of the dose absorbed into the plasma compartment was 0.78 with a time to peak concentration (4.9 μg/ml) of 8 to 12 h.
- Published
- 1999
11. Novel Approach to Specific Growth Factor Inhibition in Vivo
- Author
-
Burg M, Heinfried H. Radeke, Louis S. Green, Jürgen Floege, Stanley C. Gill, Tammo Ostendorf, Ulf Janssen, Nebojsa Janjic, and Chandra Vargeese
- Subjects
medicine.medical_specialty ,Platelet-derived growth factor ,biology ,Mesangial cell ,Aptamer ,Growth factor ,medicine.medical_treatment ,Molecular biology ,Pathology and Forensic Medicine ,Fibronectin ,Type IV collagen ,chemistry.chemical_compound ,Endocrinology ,chemistry ,In vivo ,Internal medicine ,biology.protein ,medicine ,Platelet-derived growth factor receptor - Abstract
Mesangial cell proliferation and matrix accumulation, driven by platelet-derived growth factor (PDGF), contribute to many progressive renal diseases. In a novel approach to antagonize PDGF, we investigated the effects of a nuclease-resistant high-affinity oligonucleotide aptamer in vitro and in vivo. In cultured mesangial cells, the aptamer markedly suppressed PDGF-BB but not epidermal- or fibroblast-growth-factor-2-induced proliferation. In vivo effects of the aptamer were evaluated in a rat mesangioproliferative glomerulonephritis model. Twice-daily intravenous (i.v.) injections from days 3 to 8 after disease induction of 2.2 mg/kg PDGF-B aptamer, coupled to 40-kd polyethylene glycol (PEG), led to 1) a reduction of glomerular mitoses by 64% on day 6 and by 78% on day 9, 2) a reduction of proliferating mesangial cells by 95% on day 9, 3) markedly reduced glomerular expression of endogenous PDGF B-chain, 4) reduced glomerular monocyte/macrophage influx on day 6 after disease induction, and 5) a marked reduction of glomerular extracellular matrix overproduction (as assessed by analysis of fibronectin and type IV collagen) both on the protein and mRNA level. The administration of equivalent amounts of a PEG-coupled aptamer with a scrambled sequence or PEG alone had no beneficial effect on the natural course of the disease. These data show that specific inhibition of growth factors using custom-designed, high-affinity aptamers is feasible and effective.
- Published
- 1999
12. EFFECT OF INTERMITTENT DOSING REGIMENS OF ERLOTINIB ON METHYLNITROSOUREA-INDUCED MAMMARY CARCINOGENESIS
- Author
-
Eva Szabo, Chris Tucker, Holly L. Nicastro, Vernon E. Steele, Ronald A. Lubet, M. Margaret Juliana, Stanley C. Gill, Ann M. Bode, Kenneth K. Iwata, and Clinton J. Grubbs
- Subjects
Cancer Research ,Alkylating Agents ,Blotting, Western ,Mammary Neoplasms, Animal ,Pharmacology ,Article ,Rats, Sprague-Dawley ,Erlotinib Hydrochloride ,medicine ,Animals ,Tissue Distribution ,Dosing ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Survival rate ,Dose-Response Relationship, Drug ,business.industry ,Kinase ,Cancer ,Methylnitrosourea ,medicine.disease ,Rats ,Survival Rate ,Oncology ,Toxicity ,Quinazolines ,Biomarker (medicine) ,Female ,Erlotinib ,business ,medicine.drug - Abstract
EGF receptor (EGFR) inhibitors are used in the therapy of lung and pancreatic cancers and effectively prevent cancers in multiple animal models. Although daily dosing with erlotinib is effective, weekly dosing may reduce toxicity and have advantages, particularly for prevention. We tested alternative dosing regimens for preventive/therapeutic efficacy in a rat mammary cancer model. For prevention, erlotinib was administered by gavage beginning 5 days after methylnitrosourea (MNU). For therapy and biomarker studies, rats with palpable mammary cancers were treated for six weeks or for six days, respectively. Experiment A, erlotinib (6 mg/kg body weight/day, intragastric): daily (7 times/week); one day on/one day off; and two days on/two days off. All regimens decreased tumor incidence, increased tumor latency, and decreased cancer multiplicity versus controls (P < 0.01). However, intermittent dosing was less effective than daily dosing (P < 0.05). Experiment B, erlotinib (6 mg/kg body weight/day) daily or two days on/two days off or one time per week at 42 mg/kg body weight. All regimens reduced cancer incidence and multiplicity versus controls (P < 0.01). Interestingly, daily and weekly dosing were equally effective (P > 0.5). Experiment C, erlotinib administered at 42 or 21 mg/kg body weight 1 time per week, decreased tumor incidence and multiplicity (P < 0.01). Erlotinib had a serum half-life of ≤8 hours and weekly treatment yielded effective serum levels for ≤48 hours. Daily or weekly treatment of cancer bearing rats reduced mammary tumor size 25% to 35%, whereas control cancers increased >250%. Levels of phosphorylated extracellular signal-regulated kinase (ERK) were strongly decreased in rats treated daily/weekly with erlotinib. Thus, altering the dose of erlotinib retained most of its preventive and therapeutic efficacy, and based on prior clinical studies, is likely to reduce its toxicity. Cancer Prev Res; 6(5); 448–54. ©2013 AACR.
- Published
- 2013
13. Pharmacokinetic Profile and Pharmacodynamic Effects of ASP2215, a Selective, Potent Inhibitor of FLT3/AXL, in Patients with Relapsed or Refractory Acute Myeloid Leukemia: Results from a First-in-Human Phase 1/2 Study
- Author
-
Mark J. Levis, Ogert Fisniku, Itsuro Nagase, Alexander E. Perl, Briana Sargent, Geoffrey Yuen, Jessica K. Altman, Takeshi Kadokura, Catherine C. Smith, Charles Liu, Mark R. Litzow, Stanley C. Gill, Erkut Bahceci, and Noreen Welter
- Subjects
medicine.drug_class ,business.industry ,Immunology ,Cmax ,Cell Biology ,Hematology ,Pharmacology ,Interim analysis ,Biochemistry ,Tyrosine-kinase inhibitor ,Tolerability ,Pharmacokinetics ,Oral administration ,Pharmacodynamics ,medicine ,Dosing ,business - Abstract
Introduction: ASP2215, a new tyrosine kinase inhibitor with activity against FMS-like receptor tyrosine kinase-3 (FLT3) and AXL receptor tyrosine kinase, is currently in development for the treatment of acute myeloid leukemia (AML). Methods: In an ongoing, first-in-human Phase 1/2, dose-escalation/dose-expansion study, the pharmacokinetic (PK) profile and pharmacodynamic (PD) effects of ASP2215 were evaluated under fasting conditions in patients with relapsed or refractory AML (R/R AML). Patients who met study criteria were assigned to treatment in the dose-escalation cohorts or were randomized to an open dose level in the dose-expansion cohorts. The dose-escalation cohort was a modified 3+3 with accelerated titration design that evaluated ascending oral doses of ASP2215 (20-450 mg), the dose-expansion cohort was a parallel, multi-dose-expansion cohort. Blood samples were collected and safety/tolerability assessments, including 12-lead electrocardiogram, were evaluated at protocol-specified time points for both cohorts. Effect of increasing ASP2215 exposure on inhibition of FLT3 phosphorylation, assessed via plasma inhibitory assay (PIA), was evaluated using an inhibitory PK/PD Emaxmodel. PK/PD analyses were performed to evaluate the relationship between ASP2215 exposure and changes from baseline in QTcF intervals (ΔQTcF) and changes from baseline in clinical laboratory values (e.g., creatinine kinase [ΔCK], aspartate aminotransferase [ΔAST]). Results: Data from an interim analysis of the dose-escalation/dose-expansion study were available from 215 subjects (n=23 [dose escalation], n=192 [dose expansion]). The ASP2215 PK parameters across the dose range (20-450 mg), evaluated after both single- and multiple-dose administration, are presented (Table); statistical analyses suggest the ASP2215 PK parameters are dose proportional from 20-450 mg. Median time to maximal concentration (Tmax) was observed between 2 hr and 6 hr after single and repeated oral dosing. The estimated median half-life (t1/2) ranged from approximately 45 hr to 159 hr based on the accumulation ratio; ASP2215 accumulation was extensive after multiple dose administration as reflected by the accumulation index (Rac) ranging from approximately 3.2-10. A strong correlation was shown for ASP2215 exposure-related inhibition of FLT3 phosphorylation, with >90% FLT3 inhibition observed by Day 8 at ASP2215 doses of ≥80 mg. Although a positive slope was observed between ΔQTcF and ASP2215 exposure, only 5% of the study population were reported as having a maximum post-baseline QTcF interval >500 msec. A similar trend was observed with ASP2215 concentration-related increases in ΔCK and ΔAST; however, Conclusions: ASP2215 has demonstrated exposure-related FLT3 inhibition and a pharmacokinetic profile that support once-daily oral administration for the treatment of AML in subjects who relapsed after, or are refractory to, induction or salvage treatment. Table. ASP2215 Pharmacokinetic Parameters after Multiple-Dose Administration 20 mg (n=4) 40 mg (n=3) 80 mg (n=3) 120 mg (n=3) 200 mg (n=2) 300 mg (n=3) 450 mg (n=3) Cmax(ng/mL) 45.6 (30.5, 137) 106 (76.7, 140) 396 (217, 516) 282 (248, 593) 1460 (886, 2040) 1260 (1040, 2280) 1150 (776, 1530) Tmax(hr) 4.01 (4.00, 6.00) 3.87 (0.500, 6.00) 4.33 (4.00, 4.42) 2.02 (1.95, 5.75) 6.03 (6.00, 6.07) 6.05 (4.08, 6.07) 5.00 (4.07, 5.93) AUC0-24(ng·h/mL) 926 (543, 2480) 2460 (1750, 2800) 6280 (4160, 10600) 6190 (4200, 11300) 31500 (16500, 46600) 28700 (22300, 43300) 11500 (8070, 14900) t1/2 (hr) 52.8 (39.7, 83.1) 83.7 (68.5, 243) 91.5 (60.9, 108) 45.3 (30.5, 63.3) 143 (99.7, 186) 159 (83.3, 187) 56.9 (51.5, 62.3) Rac 3.70 (2.92, 5.51) 5.55 (4.64, 15.1) 6.02 (4.18, 7.02) 3.25 (2.38, 4.33) 9.08 (6.51, 11.7) 10.1 (5.53, 11.7) 3.94 (3.62, 4.27) Data are presented as median (minimum, maximum). AUC0-24, area under the concentration-time curve between 0-24 hr; Cmax, maximal concentration; t1/2, elimination half-life; Tmax, time to maximal concentration; Rac, accumulation ratio. Disclosures Smith: Astellas: Research Funding; Plexxikon: Research Funding. Off Label Use: ASP2215 is an investigational product for the treatment of AML. Perl:Astellas US Pharma Inc.: Consultancy; Ambit/Daichi Sankyo: Consultancy; Arog Pharmaceuticals: Consultancy; Asana Biosciences: Consultancy; Actinium Pharmaceuticals: Consultancy. Altman:Novartis: Other: Advisory board; BMS: Other: Advisory board; Seattle Genetics: Other: Advisory board; Spectrum: Other: Advisory board; Ariad: Other: Advisory board; Astellas: Other: Advisory board; assistance with abstract preparation. Gill:Astellas Pharma US, Inc: Employment. Kadokura:Astellas Pharma Global Development: Employment, Other: Personal fees. Yuen:Astellas Pharma, Inc.: Employment. Fisniku:Astellas: Employment. Liu:Astellas: Employment. Nagase:Astellas: Employment. Sargent:Astellas Pharma US, Inc: Employment. Bahceci:Astellas Pharma Global Development: Employment.
- Published
- 2015
14. Escherichia coli σ70 and NusA proteins
- Author
-
P H von Hippel, T D Yager, and Stanley C. Gill
- Subjects
Small-angle X-ray scattering ,medicine.disease_cause ,Sedimentation coefficient ,chemistry.chemical_compound ,Crystallography ,Monomer ,chemistry ,Structural Biology ,Sigma factor ,Transcription (biology) ,RNA polymerase ,Transcription preinitiation complex ,medicine ,Molecular Biology ,Escherichia coli - Abstract
In this paper we examine the physical properties and potential for self-association of the Escherichia coli transcription factors, σ 70 and NusA. We show, by a combination of chemical crosslinking, equilibrium and velocity sedimentation, quasi-elastic light scattering, and small-angle X-ray scattering that NusA exists as a monomer at KCl concentrations between 0.01 and 1.5 m , and that σ 70 exists as a monomer at KCl concentrations between 0.1 and 1.5 m . The shape and hydration characteristics of each of these monomeric proteins are also examined. The results serve as background for the companion paper in which a thermodynamic analysis is made of the interactions of these transcription factors with E. coli core RNA polymerase in solution and as a component of the functional transcription complex.
- Published
- 1991
15. Escherichia coli σ70 and NusA proteins
- Author
-
Peter H. von Hippel, Stanley C. Gill, and Stephen E. Weitzel
- Subjects
Biology ,chemistry.chemical_compound ,Biochemistry ,chemistry ,Structural Biology ,Sigma factor ,Transcription (biology) ,RNA polymerase ,Transcription preinitiation complex ,Biophysics ,biology.protein ,Transcription factor II D ,Molecular Biology ,Transcription factor ,Transcription factor II B ,Polymerase - Abstract
This paper describes the binding interactions of Escherichia coli transcription factors sigma 70 and NusA with core RNA polymerase, both free in solution and as a part of the functional transcription complex. High pressure liquid chromatography gel filtration and fluorescence techniques have been used to monitor the binding of these factors to core polymerase in solution at salt concentrations roughly comparable to the in vivo environment (250 mM-KCl, 50 mM-potassium phosphate (pH 7.5]; under these conditions all the interacting species exist separately as protein monomers. We find that sigma 70 and NusA binds competitively to core polymerase with a 1:1 binding stoichiometry in this milieu, and that NusA does not bind to the polymerase holoenzyme. Association constants of approximately 2 x 10(9) and 1 x 10(7) M-1 have been measured for the sigma 70-core polymerase interaction and for the NusA-core polymerase interaction, respectively. These findings are consistent with the original formulation of the NusA-sigma 70 cycle put forward by Greenblatt & Li, and provide the basis for a further (and preliminary) quantitative examination of these same interactions within the transcription complex. We use a number of molecular biological techniques, together with data from the literature, to estimate these binding constants in various phases of the transcription cycle. In keeping with our results in solution, we find that the effective binding affinity of sigma 70 for core polymerase within the "open" promoter-polymerase complex is at least 500-fold greater than that of NusA. As the transcription complex moves from the initiation to the elongation phase these relative binding affinities are reversed; the average association constant of NusA for the core polymerase in the elongation complex remains practically the same as in free solution (approx. 3 x 10(7) M-1), while the affinity of sigma 70 for core polymerase in this complex drops to less than 5 x 10(5) M-1. These results are used to begin to define the basic conformational states and interaction potentials of core polymerase in the various stages of the transcription cycle.
- Published
- 1991
16. Preclinical and phase 1A clinical evaluation of an anti-VEGF pegylated aptamer (EYE001) for the treatment of exudative age-related macular degeneration
- Author
-
Michael L. Klein, Jerry Bill, Morton F. Goldberg, Mark Blumenkrantz, Samir C. Patel, Joan W. Miller, Dwight Henninger, Blake Tomkinson, Lawrence A. Yannuzzi, Laurie B. Wiegand, David R. Guyer, Long Shiuh Chen, Daniel F. Martin, Daniel W. Drolet, Stanley C. Gill, Denis O'Shaughnessy, Julia A. Haller, Anthony P. Adamis, Raymond A. Bendele, and Evangelos S. Gragoudas
- Subjects
Male ,Vascular Endothelial Growth Factor A ,Pathology ,Skin Neoplasms ,Angiogenesis ,Pegaptanib ,Drug Evaluation, Preclinical ,Oligonucleotides ,Endothelial Growth Factors ,Retinal Neovascularization ,chemistry.chemical_compound ,Macular Degeneration ,Mice ,Medicine ,Fluorescein Angiography ,Skin ,Anti vegf ,Aged, 80 and over ,Lymphokines ,Vascular Endothelial Growth Factors ,General Medicine ,Exudates and Transudates ,Middle Aged ,Vascular endothelial growth factor ,Female ,Rabbits ,Clinical evaluation ,medicine.drug ,Half-Life ,medicine.medical_specialty ,Aptamer ,Guinea Pigs ,Biological Availability ,Mice, Nude ,Injections ,Capillary Permeability ,Pegaptanib Sodium ,Animals ,Humans ,Corneal Neovascularization ,Retinopathy of Prematurity ,Aged ,business.industry ,Infant, Newborn ,Exudative age-related macular degeneration ,Choroidal Neovascularization ,Rats ,Vitreous Body ,Ophthalmology ,chemistry ,business - Abstract
Recent studies have suggested that vascular endothelial growth factor (VEGF) is an important stimulus for the growth of new blood vessels in the eye. Anti-VEGF therapy is thus a potential treatment for exudative macular degeneration and diabetic retinopathy.Previously described animal models of vascular leakage and ocular neovascularization, including the Miles assay, the rat corneal angiogenesis model, and the mouse retinopathy of prematurity (ROP) model, were used to study this drug. After these studies, a phase IA single ascending dose study of intravitreal injections of the drug was performed in 15 patients with subfoveal choroidal neovascularization secondary to exudative age-related macular degeneration (AMD).The Miles assay model showed almost complete attenuation of VEGF-mediated vascular leakage following addition of EYE001, and the corneal angiogenesis model also showed a significant reduction in neovascularization with EYE001. The ROP model showed inhibition of 80% of the retinal neovascularization compared with controls (P = 0.0001). The phase IA safety study of patients with exudative AMD showed no significant safety issues related to the drug. Ophthalmic evaluation revealed that 80% of patients showed stable or improved vision 3 months after treatment and that 27% of eyes demonstrated a three-line or greater improvement in vision on the Early Treatment for Diabetic Retinopathy Study chart at this time.Anti-VEGF therapy is a promising new avenue for the treatment of neovascular diseases of the eye, including exudative macular degeneration and diabetic retinopathy. Preclinical data from studies with EYE001 support clinical evaluation of its efficacy in such diseases. This report is the first to describe administration of anti-VEGF therapy in humans for exudative macular degeneration and shows the safety of such therapy for single injections. Further clinical studies are necessary to determine the safety of multiple intravitreal injections of EYE001 and larger studies are needed to prove the efficacy of this novel, potentially therapeutic agent for neovascular AMD.
- Published
- 2002
17. High-affinity aptamers selectively inhibit human nonpancreatic secretory phospholipase A2 (hnps-PLA2)
- Author
-
David W. Snyder, Philippe Bridonneau, Ying-Fon Chang, Lea M. Johnson, Theodore Goodson, David H. Parma, David K. Herron, Dan O'connell, and Stanley C. Gill
- Subjects
Male ,Indoles ,Aptamer ,Guinea Pigs ,Molecular Sequence Data ,Oligonucleotides ,In Vitro Techniques ,Group II Phospholipases A2 ,Phospholipases A ,chemistry.chemical_compound ,Phospholipase A2 ,Drug Discovery ,Animals ,Humans ,Amino Acid Sequence ,Enzyme Inhibitors ,IC50 ,Lung ,Gene Library ,biology ,Base Sequence ,Oligonucleotide ,Elastase ,Muscle, Smooth ,Molecular biology ,Phospholipases A2 ,chemistry ,Biochemistry ,Enzyme inhibitor ,Drug Design ,biology.protein ,Molecular Medicine ,Nucleic Acid Conformation ,Pleura ,RNA ,lipids (amino acids, peptides, and proteins) ,Arachidonic acid ,Systematic evolution of ligands by exponential enrichment - Abstract
A family of sequence-related 2'-aminopyrimidine, 2'-hydroxylpurine aptamers, developed by oligonucleotide-based combinatorial chemistry, SELEX (systematic evolution of ligand by exponential enrichment) technology, binds human nonpancreatic secretory phospholipase A2 (hnps-PLA2) with nanomolar affinities and inhibits enzymatic activity. Aptamer 15, derived from the family, binds hnps-PLA2 with a Kd equal to 1.7 +/- 0.2 nM and, in a standard chromogenic assay of enzymatic activity, inhibits hnps-PLA2 with an IC50 of 4 nM, at a mole fraction of substrate concentration of 4 x 10(-6) and a calculated Ki of 0.14 nM. Aptamer 15 is selective for hnps-PLA2, having a 25- and 2500-fold lower affinity, respectively, for the unrelated proteins human neutrophil elastase and human IgG. Contractions of guinea pig lung pleural strips induced by hnps-PLA2 are abolished by 0.3 microM aptamer 15, whereas contractions induced by arachidonic acid are not altered. The structure that is essential for binding and inhibition appears to be a 40-base hairpin/loop motif with an asymmetrical internal loop. The affinity and activity of the aptamers demonstrate the ability of the SELEX process to isolate antagonists of nonnucleic-acid-binding proteins from vast oligonucleotide combinatorial libraries.
- Published
- 1998
18. Nuclease-resistant nucleic acid ligands to vascular permeability factor/vascular endothelial growth factor
- Author
-
Bruce D. Feistner, Derek Jellinek, Stanley C. Gill, Louis S. Green, Laurie A. Beebe, Fiona M. Jucker, Carol Bell, and Nebojsa Janjic
- Subjects
Vascular Endothelial Growth Factor A ,in vitro evolution ,2′-amino-2′-deoxypyrimidine nucleotide RNA ,Chemical Phenomena ,Molecular Sequence Data ,Clinical Biochemistry ,Oligonucleotides ,Endothelial Growth Factors ,Biology ,Ligands ,Biochemistry ,chemistry.chemical_compound ,angiogenesis ,Ribonucleases ,combinatorial libraries ,Peptide Library ,Nucleic Acids ,Drug Discovery ,Animals ,Humans ,Nucleotide ,Nuclear Magnetic Resonance, Biomolecular ,Molecular Biology ,chemistry.chemical_classification ,Pharmacology ,Lymphokines ,Base Sequence ,Oligonucleotide ,Chemistry, Physical ,Vascular Endothelial Growth Factors ,RNA ,General Medicine ,Ligand (biochemistry) ,Molecular biology ,Recombinant Proteins ,Rats ,Vascular endothelial growth factor ,Vascular endothelial growth factor A ,chemistry ,Purines ,nuclease-resistant oligonucleotides ,Nucleic acid ,Molecular Medicine ,Systematic evolution of ligands by exponential enrichment - Abstract
Background : Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) is a potent inducer of new blood vessel growth (angiogenesis) that contributes to the pathology of many angiogenesis-associated disease states such as psoriasis, rheumatoid arthritis and cancer. Few molecular entities capable of binding to VPF/VEGF with high affinity and specificity have been described to date. Results : Nuclease-resistant 2′-amino-2′-deoxypyrimidine nucleotide RNA (2′-aminopyrimidine RNA) ligands that bind to VPF/VEGF with high affinity have been identified by iterative rounds of affinity-selection/amplification from two independent random libraries. The sequence information that confers high affinity binding toVPF/VEGF is contained in a contiguous stretch of 24 nucleotides, 5′- CCCUGAUGGUAGACGCCGGGGUG -3′ (2′-aminopyrimidine nucleotides are designated with italic letters). Of the 14 ribopurines in this minimal ligand, 10 can be substituted with the corresponding 2′-O-methylpurine nucleotides without a reduction in binding affinity to VPF/VEGF. In fact, the 2′ O-methyl substitution at permissive positions leads to a ∼17-fold improvement in the binding affinity to VPF/VEGF The higher affinity results from the reduction in the dissociation rate constant of the 2′-O-methyl-substituted RNA ligand from the protein compared to the unsubstituted ligand. The 2′-O-methyl-substituted minimal ligand, which folds into a bulged hairpin motif, is also more thermally stable than the unsubstituted ligand. Nuclease resistance of the ligand is further improved by the 2′-O-methyl substitutions and the addition of short phosphorothioate caps to the 3′- and 5′-ends. Conclusions : We have used the SELEX (systematic evolution of ligands by exponential enrichment) process in conjunction with post-SELEX modifications to define a highly nuclease-resistant oligonucleotide that binds to VPF/VEGF with high affinity and specificity.
- Published
- 1996
- Full Text
- View/download PDF
19. Oligonucleotide Ligands that Discriminate between Theophylline and Caffeine
- Author
-
Barry Polisky, Robert D. Jenison, and Stanley C. Gill
- Subjects
chemistry.chemical_compound ,chemistry ,Oligonucleotide ,medicine ,Theophylline ,Pharmacology ,Caffeine ,medicine.drug - Published
- 1995
20. Absorption, Metabolism, and Excretion of Quizartinib (AC220), a FLT3 Tyrosine Kinase Inhibitor for Treatment of Acute Myeloid Leukemia, in Healthy Male Volunteers
- Author
-
Madhu Sanga, Jianke Li, Joyce James, Tadashi Hashimoto, Gayle Bresnahan, Stanley C. Gill, Christine Hale, and Guy Gammon
- Subjects
medicine.medical_specialty ,Gastrointestinal tract ,business.industry ,Urinary system ,Metabolite ,Immunology ,Half-life ,Cell Biology ,Hematology ,Urine ,Pharmacology ,Biochemistry ,Excretion ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Internal medicine ,medicine ,business ,Feces ,Quizartinib - Abstract
Abstract 4327 Quizartinib (AC220) is an oral tyrosine kinase inhibitor that has shown promising activity in refractory/relapsed acute myeloid leukemia patients in a Phase 1 and a Phase 2 study. The absorption, metabolism, and excretion of quizartinib were characterized in healthy volunteers in a Phase 1 mass balance study. Six healthy males (mean ± SD age 29 ± 8.4 years) received a single oral dose of 60 mg quizartinib as a solution; approximately 1.6% of the total dose was labeled with 14C (approximately 100 μCi). Blood, plasma, urine, and feces were collected for 14 days (336 h) after dosing. Maximum mean ± SD blood radioactivity concentrations, reached 4 h after dosing, were 296 ± 67.4 ng equivalents/g. Maximum blood radioactivity concentrations in individual subjects occurred 4 h after dosing and ranged from 228 to 397 ng equivalents/g. Excretion of radioactivity was relatively consistent throughout the study. The maximum mean ± SD urinary concentration of radioactivity was 112 ± 52.3 ng equivalents/g at 8 to 24 hours after dosing, and the maximum mean ± SD fecal concentration of radioactivity was 51,100 ± 21,300 ng equivalents/g at 24 to 48 h after dosing. A mean ± SD of 1.64 ± 0.482% of the dose was recovered in urine, and 76.3 ± 6.23% was recovered in feces. The overall mean ± SD recovery of radioactivity in urine and feces was 78.0 ± 6.24% over the course of the study, with recovery from individual subjects ranging from 72.4% to 88.3%. Excretion of radioactivity was still ongoing at study completion (336 h). Recovery of The major radiolabeled components of plasma were unchanged quizartinib and the oxidative metabolite AC886. Five additional metabolites in plasma were identified by LC-MS but could not be identified by measurement of radioactivity, because of low levels. Eighteen radioactive peaks in urine were detected, representing less than 0.09% of the administered dose, but their putative structures could not be identified because of low levels. Quizartinib was extensively metabolized, with the metabolites excreted primarily in feces, suggesting hepatobiliary excretion of radioactivity, non-biliary excretion into the gastrointestinal tract, or metabolism within the gastrointestinal tract. Forty-two radioactive peaks were detected in fecal extracts, of which unchanged quizartinib was a significant radioactive component (mean of 4.0% of the administered radioactive dose), and 15 metabolites, each representing a mean of 1.0% to 3.5% of the administered radioactive dose, were identified. Quizartinib was predominantly metabolized by phase I biotransformations, as was evident by the absence of any conjugates of quizartinib or of oxidative metabolites under the analytical conditions used to profile plasma, urine, and feces. Quizartinib was metabolized by multiple biotransformation pathways, including oxidation, reduction, dealkylation, deamination, and hydrolysis, and by combinations of these pathways. Quizartinib was well tolerated as a single 60 mg dose in this study. There were no clinically significant changes in vital signs, ECGs, or laboratory test results. Two subjects experienced adverse events. Grade 1 dry skin in 1 subject was considered to be unrelated to study treatment, and Grade 1 diarrhea in 1 subject was considered to be possibly related to treatment. The results of this study indicated that, in humans, quizartinib was orally available and predominantly eliminated in feces, with renal clearance as a minor elimination route, and that AC886 was the only major metabolite in the circulation. Disclosures: Li: Ambit Biosciences: Employment. Bresnahan:Ambit Biosciences: Employment. Gammon:Ambit Biosciences: Employment. Sanga:Covance Laboratories, Inc.: Employment. Hale:Covance, Inc.: Employment. Hashimoto:Astellas Pharma, Inc.: Employment. Gill:Astellas Pharma, Inc.: Employment. James:Ambit Biosciences: Employment.
- Published
- 2012
21. High-resolution molecular discrimination by RNA
- Author
-
Stanley C. Gill, Barry Polisky, Arthur Pardi, and Robert D. Jenison
- Subjects
DNA, Complementary ,Magnetic Resonance Spectroscopy ,Stereochemistry ,Molecular Sequence Data ,Binding, Competitive ,chemistry.chemical_compound ,Theophylline ,medicine ,Binding site ,Binding selectivity ,Multidisciplinary ,Binding Sites ,Base Sequence ,Molecular Structure ,Chemistry ,Oligonucleotide ,RNA ,Hydrogen Bonding ,Sequence Analysis, DNA ,Ligand (biochemistry) ,Dissociation constant ,Xanthines ,Nucleic Acid Conformation ,DNA ,medicine.drug - Abstract
Species of RNA that bind with high affinity and specificity to the bronchodilator theophylline were identified by selection from an oligonucleotide library. One RNA molecule binds to theophylline with a dissociation constant Kd of 0.1 microM. This binding affinity is 10,000-fold greater than the RNA molecule's affinity for caffeine, which differs from theophylline only by a methyl group at nitrogen atom N-7. Analysis by nuclear magnetic resonance indicates that this RNA molecule undergoes a significant change in its conformation or dynamics upon theophylline binding. Binding studies of compounds chemically related to theophylline have revealed structural features required for the observed binding specificity. These results demonstrate the ability of RNA molecules to exhibit an extremely high degree of ligand recognition and discrimination.
- Published
- 1994
22. Physical properties of the Escherichia coli transcription termination factor rho. 1. Association states and geometry of the rho hexamer
- Author
-
P H von Hippel, Johannes Geiselmann, Stanley C. Gill, P. Calmettes, and T D Yager
- Subjects
Models, Molecular ,Transcription, Genetic ,Protein Conformation ,Protein subunit ,Oligonucleotides ,Neutron scattering ,Random hexamer ,Ligands ,Biochemistry ,Diffusion ,Structure-Activity Relationship ,X-Ray Diffraction ,Multienzyme Complexes ,Escherichia coli ,Scattering, Radiation ,Phase diagram ,Scattering ,Chemistry ,DNA ,Rho Factor ,Molecular Weight ,Crystallography ,Sedimentation equilibrium ,Radius of gyration ,RNA ,Protein quaternary structure ,Salts ,Ultracentrifugation ,Mathematics ,Protein Binding - Abstract
To function as a DNA-RNA helicase in rho-dependent transcript termination, six genetically identical subunits of the Escherichia coli transcription termination protein rho must first assemble into a hexameric complex. To help determine the quaternary structure of this complex, we have studied the association equilibria of the rho protomers. Sedimentation equilibrium, sedimentation velocity, diffusion, X-ray scattering, and neutron-scattering data have been combined to create a "phase diagram" of the association states of this protein as a function of protein concentration and ionic environment. The results show that rho exists predominantly as a hexamer under approximately physiological conditions and that this hexamer is in equilibrium with both lower and higher states of association that may also have physiological relevance. Small-angle X-ray scattering measurements and theoretical calculations indicate that the rho hexamer has a radius of gyration of 50 +/- 3 A. The radius of gyration measured by small-angle neutron scattering in 2H2O is 47 +/- 3 A. These scattering studies also support earlier models of rho as a planar hexagon which have been developed on the basis of electron microscopy. In the following paper in this issue [Geiselmann, J., Seifried, S. E., Yager, T. D., Liang, C.,von Hippel, P. H. (1992)], these results are combined with information on symmetry, subunit interactions, and packing geometry to obtain a model of the quaternary structure of the functional rho hexamer.
- Published
- 1992
23. Thermodynamic analysis of the transcription cycle in E. coli
- Author
-
Peter H. von Hippel, T D Yager, and Stanley C. Gill
- Subjects
Transcription, Genetic ,Protein Conformation ,Biophysics ,RNA polymerase II ,Sigma Factor ,Computational biology ,Biochemistry ,chemistry.chemical_compound ,Bacterial Proteins ,Sigma factor ,RNA polymerase ,Escherichia coli ,Promoter Regions, Genetic ,RNA polymerase II holoenzyme ,Genetics ,biology ,General transcription factor ,Models, Genetic ,Chemistry ,Organic Chemistry ,DNA-Directed RNA Polymerases ,Gene Expression Regulation, Bacterial ,Kinetics ,RNA, Bacterial ,biology.protein ,Thermodynamics ,Transcription factor II F ,Transcription factor II E ,Transcription factor II D - Abstract
The E. coli RNA transcription cycle can be divided into three major phases, which are generally called initiation, elongation, and termination. In this paper, we review recent biophysical studies of the interactions of the transcriptional regulatory proteins, sigma 70 and NusA, with themselves and with core RNA polymerase in solution, as well as with core polymerase within the transcription complex. The different affinities of sigma 70 and NusA for core RNA polymerase at various stages in the transcription cycle, together with other quantitative data, are then used to construct a partial free energy diagram for the overall transcription process. This thermodynamic framework, which is interrupted by at least two irreversible steps, can be used to rationalize physiological aspects of the transcription cycle and its regulation, as well as to identify crucial points at which our knowledge is still incomplete.
- Published
- 1990
24. Calculation of protein extinction coefficients from amino acid sequence data
- Author
-
Stanley C. Gill and P H von Hippel
- Subjects
chemistry.chemical_classification ,Chromatography ,medicine.diagnostic_test ,Globular protein ,Biophysics ,Proteins ,Cell Biology ,Molar absorptivity ,Ligand (biochemistry) ,Biochemistry ,Molecular Weight ,chemistry ,Extinction (optical mineralogy) ,Spectrophotometry ,SOSUI ,medicine ,Animals ,Humans ,Spectrophotometry, Ultraviolet ,Amino Acid Sequence ,Biological system ,Molecular Biology ,Peptide sequence ,Anticalin - Abstract
Quantitative study of protein-protein and protein-ligand interactions in solution requires accurate determination of protein concentration. Often, for proteins available only in "molecular biological" amounts, it is difficult or impossible to make an accurate experimental measurement of the molar extinction coefficient of the protein. Yet without a reliable value of this parameter, one cannot determine protein concentrations by the usual uv spectroscopic means. Fortunately, knowledge of amino acid residue sequence and promoter molecular weight (and thus also of amino acid composition) is generally available through the DNA sequence, which is usually accurately known for most such proteins. In this paper we present a method for calculating accurate (to +/- 5% in most cases) molar extinction coefficients for proteins at 280 nm, simply from knowledge of the amino acid composition. The method is calibrated against 18 "normal" globular proteins whose molar extinction coefficients are accurately known, and the assumptions underlying the method, as well as its limitations, are discussed.
- Published
- 1989
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.