Network


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

Hotspot


Dive into the research topics where Howard H. Bailey is active.

Publication


Featured researches published by Howard H. Bailey.


Journal of Biological Chemistry | 1997

Constitutive and β-Naphthoflavone-induced Expression of the Human γ-Glutamylcysteine Synthetase Heavy Subunit Gene Is Regulated by a Distal Antioxidant Response Element/TRE Sequence

Mulcahy Rt; Wartman Ma; Howard H. Bailey; Gipp Jj

Glutathione (GSH) is an abundant cellular non-protein sulfhydryl that functions as an important protectant against reactive oxygen species and electrophiles, is involved in the detoxification of xenobiotics, and contributes to the maintenance of cellular redox balance. The rate-limiting enzyme in the de novo synthesis of glutathione is γ-glutamylcysteine synthetase (GCS), a heterodimer consisting of heavy and light subunits expressing catalytic and regulatory functions, respectively. Exposure of HepG2 cells to β-naphthoflavone (β-NF) resulted in a time- and dose-dependent increase in the steady-state mRNA levels for both subunits. In order to identify sequences mediating the constitutive and induced expression of the heavy subunit gene, a series of deletion mutants created from the 5′-flanking region (−3802 to +465) were cloned into a luciferase reporter vector (pGL3-Basic) and transfected into HepG2 cells. Constitutive expression was maximally directed by sequences between −202 and +22 as well as by elements between −3802 to −2752. The former sequence contains a consensus TATA box. Increased luciferase expression following exposure to 10 μM β-NF was only detected in cells transfected with a reporter vector containing the full-length −3802:+465 fragment. Hence, elements directing constitutive and induced expression of the GCS heavy subunit are present in the distal portion of the 5′-flanking region, between positions −3802 and −2752. Sequence analysis revealed the presence of several putative consensus response elements in this region, including two potential antioxidant response elements (ARE3 and ARE4), separated by 34 base pairs. When cloned into the thymidine kinase-luciferase vector, pT81-luciferase, and transfected into HepG2 cells, both ARE3 and ARE4 increased basal luciferase expression approximately 20-fold. When cloned in tandem in their native arrangement the increase in luciferase activity was in excess of 100-fold, suggesting a strong interaction between the two sequences. Luciferase expression was elevated in β-NF-treated cells transfected with the ARE4-tk-luciferase vector and all DNA fragments containing ARE4. In contrast, ARE3 did not direct increased luciferase expression in response to β-NF nor did it significantly modify the magnitude of induction directed by ARE4. The influence of the ARE4 oligonucleotide on constitutive and induced expression was eliminated by introduction of a single base mutation, converting the core ARE sequence in ARE4 from 5′-GGACTCAGCG-3′ to 5′-GGACTCAGCG-3′. When introduced into the full-length −3802:+465 segment, the same single base mutation also eliminated both functions. Collectively the data indicate that the constitutive and β-NF-induced expression of the human GCS heavy subunit gene is mediated by a distal ARE sequence containing an embedded tetradecanoylphorbol-13-acetate-responsive element.


Journal of the National Cancer Institute | 2010

Chemoprevention of Nonmelanoma Skin Cancer With Celecoxib: A Randomized, Double-Blind, Placebo-Controlled Trial

Craig A. Elmets; Jaye L. Viner; Alice P. Pentland; Wendy Cantrell; Hui Yi Lin; Howard H. Bailey; Sewon Kang; Kenneth G. Linden; Michael P. Heffernan; Madeleine Duvic; Ellen Richmond; Boni E. Elewski; Asad Umar; Walter C. Bell; Gary B. Gordon

Background Preclinical studies indicate that the enzyme cyclooxygenase 2 plays an important role in ultraviolet-induced skin cancers. We evaluated the efficacy and safety of celecoxib, a cyclooxygenase 2 inhibitor, as a chemopreventive agent for actinic keratoses, the premalignant precursor of nonmelanoma skin cancers, and for nonmelanoma skin cancers, including cutaneous squamous cell carcinomas (SCCs) and basal cell carcinomas (BCCs). Methods A double-blind placebo-controlled randomized trial involving 240 subjects aged 37–87 years with 10–40 actinic keratoses was conducted at eight US academic medical centers. Patients were randomly assigned to receive 200 mg of celecoxib or placebo administered orally twice daily for 9 months. Subjects were evaluated at 3, 6, 9 (ie, completion of treatment), and 11 months after randomization. The primary endpoint was the number of new actinic keratoses at the 9-month visit as a percentage of the number at the time of randomization. In an intent-to-treat analysis, the incidence of actinic keratoses was compared between the two groups using t tests. In exploratory analyses, we evaluated the number of nonmelanoma skin cancers combined and SCCs and BCCs separately per patient at 11 months after randomization using Poisson regression, after adjustment for patient characteristics and time on study. The numbers of adverse events in the two treatment arms were compared using χ2 or Fisher exact tests. All statistical tests were two-sided. Results There was no difference in the incidence of actinic keratoses between the two groups at 9 months after randomization. However, at 11 months after randomization, there were fewer nonmelanoma skin cancers in the celecoxib arm than in the placebo arm (mean cumulative tumor number per patient 0.14 vs 0.35; rate ratio [RR] = .43, 95% confidence interval [CI] = 0.24 to 0.75; P = .003). After adjusting for age, sex, Fitzpatrick skin type, history of actinic keratosis at randomization, nonmelanoma skin cancer history, and patient time on study, the number of nonmelanoma skin cancers was lower in the celecoxib arm than in the placebo arm (RR = 0.41, 95% CI = 0.23 to 0.72, P = .002) as were the numbers of BCCs (RR = 0.40, 95% CI = 0.18 to 0.93, P = .032) and SCCs (RR = 0.42, 95% CI = 0.19 to 0.93, P = .032). Serious and cardiovascular adverse events were similar in the two groups. Conclusions Celecoxib may be effective for prevention of SCCs and BCCs in individuals who have extensive actinic damage and are at high risk for development of nonmelanoma skin cancers.


Journal of Clinical Oncology | 1994

Phase I clinical trial of intravenous L-buthionine sulfoximine and melphalan: An attempt at modulation of glutathione

Howard H. Bailey; R T Mulcahy; Kendra D. Tutsch; Rhoda Z. Arzoomanian; D. Alberti; Mary B. Tombes; George Wilding; Marcia Pomplun; David Spriggs

PURPOSE A phase I dose-escalation trial of intravenous (IV) L-buthionine-SR-sulfoximine (BSO) with melphalan (L-PAM) was performed to determine the toxicity and biologic activity of BSO, administered as a short infusion every 12 hours, and the combination of BSO plus L-PAM. PATIENTS AND METHODS Twenty-eight patients with refractory malignancies received 30-minute infusions of BSO every 12 hours for 6 to 10 doses in week 1 followed in week 2 by either IV L-PAM (15 mg/m2) alone or BSO as in week 1 with L-PAM. Patients received the combination in week 5 (course no. 2) if they received L-PAM alone during week 2 and vice versa. BSO doses ranged from 1.5 g/m2 to 13.104 g/m2. RESULTS The only toxicity observed with BSO infusions was occasional nausea/vomiting. Evaluation of 23 paired courses (L-PAM plus BSO v L-PAM) showed significantly (P < .001) greater leukopenia and thrombocytopenia with L-PAM plus BSO. No other significant toxicity was noted. Measurement of intracellular glutathione (GSH) levels in peripheral mononuclear cells (PBLs) of all patients receiving BSO showed a consistent, non-dose-dependent, linear decrease in GSH with repeated BSO doses. Maximal GSH depletion (40% of baseline values, absolute values 200 to 250 ng/10(6) PBLs) was noted after the sixth BSO dose; extended BSO dosing schedules beyond six total BSO doses did not further deplete GSH. Evaluation of gamma-glutamylcysteine synthetase (GCS) activity showed marked inhibition near the end of each infusion with near complete recovery of GCS activity before the next BSO dose. The pattern of GCS inhibition mirrored the plasma BSO concentrations with peak values (level 6, 4 to 8 mmol/L L,R+L,S BSO) observed at the end of the infusion with a rapid decrease in plasma concentrations with an estimated half-life (t1/2) of less than 2 hours. Differential elimination of the R+S stereoisomers was observed. Analysis of L-PAM pharmacokinetics showed marked interpatient variability and a significant decrease in total-body clearance (P = .01) and volume of distribution (P = .03) in courses with L-PAM plus BSO as compared with L-PAM alone. CONCLUSION This study shows that BSO alone and in combination with L-PAM can be safely given to patients, but that a schedule of short infusions every 12 hours does not result in GSH depletion less than 30% of baseline values.


Phytomedicine | 2010

Green tea polyphenols for prostate cancer chemoprevention: a translational perspective.

Jeremy J. Johnson; Howard H. Bailey; Hasan Mukhtar

Every year nearly 200,000 men in the United States are diagnosed with prostate cancer (PCa), and another 29,000 men succumb to the disease. Within certain regions of the world population based studies have identified a possible role for green tea in the prevention of certain cancers, especially PCa. One constituent in particular, epigallocatechin-3-gallate also known as EGCG has been shown in cell culture models to decrease cell viability and promote apoptosis in multiple cancer cell lines including PCa with no effect on non-cancerous cell lines. In addition, animal models have consistently shown that standardized green tea polyphenols when administered in drinking water delay the development and progression of PCa. Altogether, three clinical trials have been performed in PCa patients and suggest that green tea may have a distinct role as a chemopreventive agent. This review will present the available data for standardized green tea polyphenols in regard to PCa chemoprevention that will include epidemiological, mechanism based studies, safety, pharmacokinetics, and applicable clinical trials. The data that has been collected so far suggests that green tea may be a promising agent for PCa chemoprevention and further clinical trials of participants at risk of PCa or early stage PCa are warranted.


Chemico-Biological Interactions | 1998

l-S,R-buthionine sulfoximine: historical development and clinical issues

Howard H. Bailey

L-S,R-buthionine sulfoximine (L-S,R BSO) is a potent specific inhibitor of gamma-glutamylcysteine synthetase, the rate-limiting step in glutathione (GSH) biosynthesis. GSH is an important component of tumor drug resistance based on a strong association and recent transfection studies. Depletion of intracellular GSH by BSO significantly enhances the cytotoxicity of many cytotoxic agents, principally alkylating agents and platinating compounds but also irradiation and anthracyclines. Phase I clinical trials of BSO + melphalan (L-PAM)have been carried out and observed little toxicity with BSO alone and increased myelosuppression with BSO + L-PAM. Consistent and profound (< 10% of control) GSH depletion was observed in serial determinations of tumor GSH levels in patients receiving continuous infusion (CI) BSO. Evidence of clinical activity has been observed in patients with alkylating or platinating agent-refractory tumors. Phase II evaluation of CI BSO with L-PAM is in progress.


Cancer Chemotherapy and Pharmacology | 1994

Human metabolism of the experimental cancer therapeutic agentd-limonene

Pamela L. Crowell; Charles E. Elson; Howard H. Bailey; Abiodun Elegbede; Jill D. Haag; Michael N. Gould

Abstractd-Limonene has efficacy in preclinical models of breast cancer, causing >80% of carcinomas to regress with little host toxicity. We performed a pilot study on healthy human volunteers to identify plasma metabolites of limonene and to assess the toxicity of supradietary quantities ofd-limonene. Seven subjects ingested 100 mg/kg limonene in a custard. Blood was drawn at 0 and 24 h for chemistry-panel analysis and at 0, 4, and 24 h for limonenemetabolite analysis. On-line capillary gas chromatography/mass spectrometry (GC/MS) analysis indicated that at least five compounds were present at 4 h that were not present at time zero. Two major peaks were identified as the rat limonene metabolites dihydroperillic acid and perillic acid, and two minor peaks were found to be the respective methyl esters of these acids. A third major peak was identified as limonene-1,2-diol. Limonene was a minor component. At a dose of 100 mg/kg, limonene caused no gradable toxicity. Limonene is metabolized by humans and rats in a similar manner. These observations and the high therapeutic ratio of limonene in the chemotherapy of rodent cancers suggest that limonene may be an efficacious chemotherapeutic agent for human malignancies.


Cancer | 2006

Phase II study of daily oral perifosine in patients with advanced soft tissue sarcoma

Howard H. Bailey; Michelle R. Mahoney; David S. Ettinger; William J. Maples; Paula M. Fracasso; Anne M. Traynor; Charles Erlichman; Scott H. Okuno

A multicenter Phase II study was performed to evaluate the clinical activity of an initial loading (150 mg every 6 hours × 4 doses) dose followed by continuous daily oral dosing (100 mg/day) of perifosine in patients with advanced soft tissue sarcomas (STSs).


Cancer | 2011

A Phase 2 Study of Temsirolimus (CCI-779) in Patients With Soft Tissue Sarcomas: A Study of the Mayo Phase 2 Consortium (P2C)

Scott H. Okuno; Howard H. Bailey; Michelle R. Mahoney; Douglas Adkins; William J. Maples; Tom R. Fitch; David S. Ettinger; Charles Erlichman; Jann N. Sarkaria

The primary goal of this trial was to evaluate the confirmed response rate of temsirolimus (CCI‐779), a mammalian target of rapamycin in patients with advanced soft tissue sarcomas (STS).


Cancer Chemotherapy and Pharmacology | 1994

Up-regulation of γ-glutamylcysteine synthetase activity in melphalan-resistant human multiple myeloma cells expressing increased glutathione levels

R. Timothy Mulcahy; Howard H. Bailey; Jerry J. Gipp

Levels of intracellular glutathione (GSH) and the GSH-related enzymes γ-glutamylcysteine synthetase (γ-GCS) and γ-glutamyltranspeptidase (γ-GT) were measured in the melphalan-resistant human multiple myeloma cell line 8226/LR-5 and were compared to those measured in the drug-sensitive 8226/S and doxorubicin-resistant 8226/Dox40 cell lines. Both GSH and γ-GCS activity, the rate-limiting step in the de novo synthesis of GSH, were elevated by a factor of approximately 2 in the melphalanresistant 8226/LR-5 cells relative to the other two lines. γ-GT activity was not elevated significantly in the /LR-5 cells. Northern analysis with a probe specific for the large subunit of human liver γ-GCS identified two bands (3.2 and 4.0 kb), both of which were increased by a factor of 2–3 in the 8226/LR-5 line. Levels of γ-GCS mRNA expression were comparable in the /S and /Dox40 cell lines. Levels of γ-GT mRNA were similar in the /S and /LR-5 lines but were reduced in the /Dox40 cells. These data suggest that the increased GSH levels associated with resistance to melphalan in the 8226/LR-5 myeloma cells is attributable to up-regulation of γ-GCS. This observation is consistent with recent demonstrations of up-regulation of γ-GCS in melphalan-resistant prostate carcinoma cells and cisplatinum-resistant ovarian carcinoma cells, suggesting that increased expression of γ-GCS may be an important mediator of GSH-associated resistance mechanisms.


Cancer Prevention Research | 2010

A randomized, double-blind, placebo-controlled phase 3 skin cancer prevention study of {alpha}-difluoromethylornithine in subjects with previous history of skin cancer.

Howard H. Bailey; KyungMann Kim; Ajit K. Verma; Karen Sielaff; Paul O. Larson; Stephen N. Snow; T Lenaghan; Jaye L. Viner; Jeff Douglas; Nancy E. Dreckschmidt; Mary Hamielec; Marcia Pomplun; Harry Sharata; Puchalsky D; Er Berg; Thomas C. Havighurst; Paul P. Carbone

Preclinical studies have shown that the inhibition of ornithine decarboxylase (ODC) by α-difluoromethylornithine (DFMO) and resultant decreases in tissue concentrations of polyamines (putrescine and spermidine) prevents neoplastic developments in many tissue types. Clinical studies of oral DFMO at 500 mg/m2/day revealed it to be safe and tolerable and resulted in significant inhibition of phorbol ester–induced skin ODC activity. Two hundred and ninety-one participants (mean age, 61 years; 60% male) with a history of prior nonmelanoma skin cancer (NMSC; mean, 4.5 skin cancers) were randomized to oral DFMO (500 mg/m2/day) or placebo for 4 to 5 years. There was a trend toward a history of more prior skin cancers in subjects randomized to placebo, but all other characteristics including sunscreen and nonsteroidal anti-inflammatory drug use were evenly distributed. Evaluation of 1,200 person-years of follow-up revealed a new NMSC rate of 0.5 events/person/year. The primary end point, new NMSCs, was not significantly different between subjects taking DFMO and placebo (260 versus 363 cancers, P = 0.069, two-sample t test). Evaluation of basal cell (BCC) and squamous cell cancers separately revealed very little difference in squamous cell cancer between treatment groups but a significant difference in new BCC (DFMO, 163 cancers; placebo, 243 cancers; expressed as event rate of 0.28 BCC/person/year versus 0.40 BCC/person/year, P = 0.03). Compliance with DFMO was >90% and it seemed to be well tolerated with evidence of mild ototoxicity as measured by serial audiometric examination when compared with placebo subjects. The analysis of normal skin biopsies revealed a significant (P < 0.05) decrease in 12-0-tetradecanoylphorbol-13-acetate–induced ODC activity (month 24, 36, and 48) and putrescine concentration (month 24 and 36 only) in DFMO subjects. Subjects with a history of skin cancer taking daily DFMO had an insignificant reduction (P = 0.069) in new NMSC that was predominantly due to a marked reduction in new BCC. Based on these data, the potential of DFMO, alone or in combination, to prevent skin cancers should be explored further. Cancer Prev Res; 3(1); 35–47

Collaboration


Dive into the Howard H. Bailey's collaboration.

Top Co-Authors

Avatar

George Wilding

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Dona Alberti

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

KyungMann Kim

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jill M. Kolesar

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Kendra D. Tutsch

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Rhoda Z. Arzoomanian

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jens C. Eickhoff

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Rebecca Marnocha

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Glenn Liu

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Thomas C. Havighurst

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge