Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Scott N. Holden is active.

Publication


Featured researches published by Scott N. Holden.


Clinical Cancer Research | 2006

A Phase I Biological and Pharmacologic Study of the Heparanase Inhibitor PI-88 in Patients with Advanced Solid Tumors

Michele Basche; Daniel L. Gustafson; Scott N. Holden; Cindy L. O'Bryant; Lia Gore; Samir E. Witta; Mary Kay Schultz; Mark Morrow; Adrah Levin; Brian R. Creese; Michael Kangas; Kaye L. Roberts; Thu Suong T. Nguyen; Kat Davis; R. S. Addison; Jane C. Moore; S. Gail Eckhardt

Purpose: PI-88 is a mixture of highly sulfated oligosaccharides that inhibits heparanase, an extracellular matrix endoglycosidase, and the binding of angiogenic growth factors to heparan sulfate. This agent showed potent inhibition of placental blood vessel angiogenesis as well as growth inhibition in multiple xenograft models, thus forming the basis for this study. Experimental Design: This study evaluated the toxicity and pharmacokinetics of PI-88 (80-315 mg) when administered s.c. daily for 4 consecutive days bimonthly (part 1) or weekly (part 2). Results: Forty-two patients [median age, 53 years (range, 19-78 years); median performance status, 1] with a range of advanced solid tumors received a total of 232 courses. The maximum tolerated dose was 250 mg/d. Dose-limiting toxicity consisted of thrombocytopenia and pulmonary embolism. Other toxicity was generally mild and included prolongation of the activated partial thromboplastin time and injection site echymosis. The pharmacokinetics were linear with dose. Intrapatient variability was low and interpatient variability was moderate. Both AUC and Cmax correlated with the percent increase in activated partial thromboplastin time, showing that this pharmacodynamic end point can be used as a surrogate for drug exposure. No association between PI-88 administration and vascular endothelial growth factor or basic fibroblast growth factor levels was observed. One patient with melanoma had a partial response, which was maintained for >50 months, and 9 patients had stable disease for ≥6 months. Conclusion: The recommended dose of PI-88 administered for 4 consecutive days bimonthly or weekly is 250 mg/d. PI-88 was generally well tolerated. Evidence of efficacy in melanoma supports further evaluation of PI-88 in phase II trials.


Anti-Cancer Drugs | 2008

A phase I safety, tolerability, and pharmacokinetic study of enzastaurin combined with capecitabine in patients with advanced solid tumors

D. Ross Camidge; S. Gail Eckhardt; Lia Gore; Cindy L. OʼBryant; Stephen Leong; Michelle Basche; Scott N. Holden; Luna Musib; John Baldwin; Christelle Darstein; Donald Thornton; Richard S. Finn; Carolyn D. Britten

Enzastaurin, an oral inhibitor of protein kinase C&bgr;, affects signal transduction associated with angiogenesis, proliferation, and survival. Capecitabine is converted to 5-fluoruracil by thymidine phosphorylase, a putative angiogenic factor. The all-oral combination of the two drugs offers the potential for targeting angiogenesis in capecitabine-sensitive tumors with nonoverlapping toxicities. Patients with advanced cancer initially received single-agent enzastaurin to achieve steady-state concentrations (cycle 1). In subsequent 21-day cycles, enzastaurin was given orally, once daily, on days 1–21 and capecitabine orally, twice daily (b.i.d.), on days 1–14 in three dose-level cohorts. Three dose-escalation cohorts were studied: cohort 1 (n=8), 350 mg of enzastaurin +capecitabine (750 mg/m2 b.i.d.); cohort 2 (n=7), enzastaurin (350 mg)+capecitabine (1000 mg/m2 b.i.d.); cohort 3 (n=12), 525-mg capsules or 500-mg enzastaurin+capecitabine (1000 mg/m2 b.i.d.). Further dose escalation was not pursued because of emerging data that enzastaurin systemic exposure did not increase at doses above 525 mg. Although a traditional toxicity-based maximum tolerated dose was not achieved, the highest dosing cohort represented a biologically relevant dose of enzastaurin, on the basis of preclinical data and correlative pharmacodynamic biomarker assays of protein kinase C&bgr; inhibition in peripheral blood mononucleocytes, in combination with a standard dose of capecitabine. For the 500/525-mg dose, ratios of total enzastaurin analyte geometric means (i.e. enzastaurin alone versus enzastaurin with capecitabine) reflected a trend toward decreased enzastaurin exposure, but did not reach statistical significance. The pharmacokinetic parameters of capecitabine with enzastaurin were similar to those previously reported for single-agent capecitabine. The regimen was well tolerated, without any consistent pattern of drug-related grade 3 or grade 4 toxicities being observed. Although no objective tumor responses were documented, five patients maintained stable disease for ≥6 months (range: 6–9.7 months). The recommended phase II dose of this combination, based on the results of this study, is enzastaurin at a daily dose of 500 mg (tablet formulation) and capecitabine (1000 mg/m2, b.i.d.) on days 1–14 every 21 days. Further disease-directed studies are warranted, such as in malignancies in the treatment of which both capecitabine and inhibitors of angiogenesis have previously been benchmarked as being effective.


Clinical Cancer Research | 2004

A Phase I and Pharmacokinetic Study of Exisulind and Docetaxel in Patients with Advanced Solid Tumors

Samir E. Witta; Daniel L. Gustafson; A. Scott Pierson; Alexander Menter; Scott N. Holden; Michele Basche; Martha Persky; Cindy L. O'Bryant; Chan Zeng; Anna E. Barón; Michael E. Long; Amy Gibbs; Karen Kelly; Paul A. Bunn; Daniel C. Chan; Patrick Pallansch; S. Gail Eckhardt

Purpose: Exisulind (sulindac sulfone, FGN-1, Aptosyn) is a sulindac metabolite that induces apoptosis via inhibition of cyclic GMP-phosphodiesterase. This agent demonstrated tumor growth inhibition in rodent models of colon, breast, prostate, and lung carcinogenesis. In an orthotopic model of human non–small-cell lung cancer, the combination of exisulind and docetaxel prolonged survival in athymic nude rats, forming the basis of this phase I combination study. Experimental Design: This study evaluated the toxicity and pharmacokinetics of combining exisulind (150–250 mg) given orally twice daily and docetaxel (30–36 mg/m2) administered intravenously on days 1, 8, and 15 of a 4-week cycle. Results: Twenty patients with a range of advanced solid tumors (median age, 59 years; age range, 35–77 years; median performance status, 1) received a total of 70 courses. Observed adverse events were mild to moderate, and there was no dose-limiting toxicity at any level. Grade 3 gastrointestinal toxicities were present in 10 of the 70 cycles (10%) and included nausea, vomiting, dyspepsia, and elevated alkaline phosphatase. Neutropenia was present in four cycles in patients treated with a docetaxel dose of 36 mg/m2. Pharmacokinetic analysis did not demonstrate a clear effect of exisulind on docetaxel pharmacokinetics and vice versa. Relationships were evident between the plasma concentration of exisulind and the development of grade 2 or greater toxicities. One third of patients maintained stable disease for 3 to 12 cycles, but no objective responses were observed. Conclusions: The combination of docetaxel (36 mg/m2, weekly) and exisulind (500 mg/d) was reasonably well tolerated, and it is undergoing phase II testing in patients with non–small-cell lung cancer.


Journal of Clinical Oncology | 2004

Updated results from a phase I trial of the histone deacetylase (HDAC) inhibitor MS-275 in patients with refractory solid tumors

Lia Gore; Scott N. Holden; M. Basche; S. Raj; I. Arnold; Cindy L. O'Bryant; S. Witta; B. Rohde; C. McCoy; S. G. Eckhardt

3026 Background: Inhibition of HDACs in vitro has resulted in tumor cell growth arrest, differentiation and/or tumor apoptosis. This report evaluates the pharmacokinetics (PK) and pharmacodynamics (PD) of MS-275, an orally active synthetic benzamide derivative HDAC inhibitor, and presents updated clinical data from this phase I trial. METHODS MS-275 was given orally to 3-6 fasting patients per dose level with refractory/relapsed solid tumors or lymphoma. The plasma PK profile of MS-275 was analyzed using a validated, quantitative method. Histone H3 and H4 acetylation was analyzed in peripheral blood mononuclear cells (PBMCs) by immunohistochemical detection. RESULTS Seventeen pts have been enrolled on 3 schedules (Schs): 2-6 mg/m2 biweekly (n=10, Sch A); 2 mg/m2 twice weekly for 3 weeks with 1 week rest (n=6, Sch B); 4 mg/m2 weekly for 3 weeks with 1 week rest (n=1, Sch C). Eighty cycles have been administered on Sch A and 13 on Sch B. No drug related grade (gr) 4 adverse events (AEs) have been reported. The most common drug related AEs were gr 1-3 hypophosphatemia, asthenia, nausea and anorexia. The maximum tolerated dose was not reached on Sch A. Dose escalation of Sch B was not pursued due to laboratory abnormalities resulting in dose delays/omissions. The plasma profile of MS-275 demonstrates rapid absorption, with a Tmax of 0.5-2.0 h, and a dose-dependent increase in systemic exposure over the dose range 2-6 mg/m2. A biphasic elimination was noted, with an estimated T1/2 of 100 hours. Preliminary PD analyses indicate an increase in histone H3 and H4 acetylation in PBMCs, compared with pretreatment. One pt with melanoma continued to exhibit a partial response (Sch A), and 1 pt each with Ewings sarcoma (Sch A), rectal carcinoma (Sch A) and melanoma (Sch B) had stable disease after 60+, 38+ and 20+ weeks of therapy, respectively. CONCLUSIONS PK analyses indicate that MS-275 is rapidly absorbed, with a T1/2 of 100 h, and preliminary evidence of increased H3 and H4 histone acetylation has been observed. Potential antitumor activity has been noted in 4 pts and enrollment on MS-275 4 mg/m2 weekly for 3 weeks with 1 week rest is ongoing. [Table: see text].


Molecular Cancer Therapeutics | 2008

A phase I study of gefitinib, capecitabine, and celecoxib in patients with advanced solid tumors

Elaine T. Lam; Cindy L. O'Bryant; Michele Basche; Daniel L. Gustafson; Natalie J. Serkova; Baron A; Scott N. Holden; Dancey J; S. G. Eckhardt; Lia Gore

This phase I study was designed to determine the maximum tolerated dose (MTD) and toxicity profile of the combination of gefitinib, capecitabine, and celecoxib in patients with advanced solid tumors. Patients were treated with escalating doses of gefitinib once daily, capecitabine twice daily (14 of 28 days), and celecoxib twice daily. Plasma samples for biomarkers were obtained at baseline and weekly for the first 2 cycles. Pharmacokinetic variables were correlated with toxicity and presence of biological effect. Tumor biopsies from 5 patients were analyzed for changes in tumor metabolic activity by nuclear magnetic resonance spectroscopy. [18F]fluorodeoxyglucose positron emission tomography was done as a correlate in 6 patients at the MTD. Thirty-nine patients received 168 cycles of therapy. The dose-limiting toxicities observed included nausea, dehydration and nausea, diarrhea, and stomatitis. The MTD was 250 mg/d gefitinib (days 1-14) and 2,000 mg/m2/d capecitabine divided twice daily (days 8-21) every 28 days. Celecoxib was eliminated due to concerns of increased risk for cardiovascular toxicity, although no patients in this study had cardiac events. One patient with cholangiocarcinoma had a confirmed partial response. Fourteen of 39 (36%) patients maintained prolonged stable disease for a median of 4 months (range, 3-24 months). [18F]fluorodeoxyglucose positron emission tomography scan and metabolomic analyses revealed differences in metabolic response to gefitinib versus capecitabine. The combination of gefitinib and capecitabine is well tolerated and appears to have activity against certain advanced solid tumors, providing a rationale for further evaluation in advanced solid malignancies. [Mol Cancer Ther 2008;7(12):3685–94]


Cancer Chemotherapy and Pharmacology | 2003

Analysis of docetaxel pharmacokinetics in humans with the inclusion of later sampling time-points afforded by the use of a sensitive tandem LCMS assay.

Daniel L. Gustafson; Michael E. Long; Joseph A. Zirrolli; Mark W. Duncan; Scott N. Holden; A. Scott Pierson; S. Gail Eckhardt


Seminars in Oncology | 2002

Angiogenesis inhibitors in clinical development for lung cancer.

Roy S. Herbst; Manuel Hidalgo; A. Scott Pierson; Scott N. Holden; Michael Bergen; S. Gail Eckhardt


Cancer Chemotherapy and Pharmacology | 2005

Safety and pharmacokinetic study of RPI.4610 (ANGIOZYME), an anti-VEGFR-1 ribozyme, in combination with carboplatin and paclitaxel in patients with advanced solid tumors

Hiroyuki Kobayashi; S. Gail Eckhardt; Jennifer A. Lockridge; Mace L. Rothenberg; Alan Sandler; Cindy L. O’Bryant; Wendy Cooper; Scott N. Holden; Roger D. Aitchison; Nassim Usman; Maurice J. Wolin; Michele Basche


Cancer Chemotherapy and Pharmacology | 2008

A phase I pharmacological and biological study of PI-88 and docetaxel in patients with advanced malignancies

Laura Quan Man Chow; Daniel L. Gustafson; Cindy L. O’Bryant; Lia Gore; Michele Basche; Scott N. Holden; Mark Morrow; Stacy Grolnic; Brian R. Creese; Kaye L. Roberts; Kat Davis; R. S. Addison; S. Gail Eckhardt


Journal of Clinical Oncology | 2017

Human pharmacokinetic (PK) characterization of the novel dual-action anti-HER3/EGFR antibody MEHD7945A (MEHD) in patients with refractory/recurrent epithelial tumors.

Manuel Hidalgo; Antonio Calles; Dejan Juric; Rodrigo Dienstmann; Desamparados Roda Perez; Wells A. Messersmith; George R. Blumenschein; José Baselga; Josep Tabernero; Andres Cervantes-Ruiperez; Antonio Jimeno; Denise Jin; Shuang Bai; Pavel Gurevich; Ihsan Nijem; Scott N. Holden; John D. Davis; Wendy G. Halpern; Andrea Pirzkall; Yan Xin

Collaboration


Dive into the Scott N. Holden's collaboration.

Top Co-Authors

Avatar

S. Gail Eckhardt

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lia Gore

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Cindy L. O'Bryant

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Michele Basche

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

A. Scott Pierson

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Mark Morrow

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Cindy L. O’Bryant

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Manuel Hidalgo

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge