Network


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

Hotspot


Dive into the research topics where Chad A. Walden is active.

Publication


Featured researches published by Chad A. Walden.


PLOS ONE | 2011

Modulating Pharmacokinetics, Tumor Uptake and Biodistribution by Engineered Nanoparticles

Rochelle R. Arvizo; Oscar R. Miranda; Daniel F. Moyano; Chad A. Walden; Karuna Giri; Resham Bhattacharya; J. David Robertson; Vincent M. Rotello; Joel M. Reid; Priyabrata Mukherjee

Background Inorganic nanoparticles provide promising tools for biomedical applications including detection, diagnosis and therapy. While surface properties such as charge are expected to play an important role in their in vivo behavior, very little is known how the surface chemistry of nanoparticles influences their pharmacokinetics, tumor uptake, and biodistribution. Method/Principal Findings Using a family of structurally homologous nanoparticles we have investigated how pharmacological properties including tumor uptake and biodistribution are influenced by surface charge using neutral (TEGOH), zwitterionic (Tzwit), negative (TCOOH) and positive (TTMA) nanoparticles. Nanoparticles were injected into mice (normal and athymic) either in the tail vein or into the peritoneum. Conclusion Neutral and zwitterionic nanoparticles demonstrated longer circulation time via both IP and IV administration, whereas negatively and positively charged nanoparticles possessed relatively short half-lives. These pharmacological characteristics were reflected on the tumor uptake and biodistribution of the respective nanoparticles, with enhanced tumor uptake by neutral and zwitterionic nanoparticles via passive targeting.


PLOS ONE | 2013

Inhibiting the growth of pancreatic adenocarcinoma in vitro and in vivo through targeted treatment with designer gold nanotherapeutics.

Rachel A. Kudgus; Annamária Szabolcs; Jameel Ahmad Khan; Chad A. Walden; Joel M. Reid; J. David Robertson; Resham Bhattacharya; Priyabrata Mukherjee

Background Pancreatic cancer is one of the deadliest of all human malignancies with limited options for therapy. Here, we report the development of an optimized targeted drug delivery system to inhibit advanced stage pancreatic tumor growth in an orthotopic mouse model. Method/Principal Findings Targeting specificity in vitro was confirmed by preincubation of the pancreatic cancer cells with C225 as well as Nitrobenzylthioinosine (NBMPR - nucleoside transporter (NT) inhibitor). Upon nanoconjugation functional activity of gemcitabine was retained as tested using a thymidine incorporation assay. Significant stability of the nanoconjugates was maintained, with only 12% release of gemcitabine over a 24-hour period in mouse plasma. Finally, an in vivo study demonstrated the inhibition of tumor growth through targeted delivery of a low dose of gemcitabine in an orthotopic model of pancreatic cancer, mimicking an advanced stage of the disease. Conclusion We demonstrated in this study that the gold nanoparticle-based therapeutic containing gemcitabine inhibited tumor growth in an advanced stage of the disease in an orthotopic model of pancreatic cancer. Future work would focus on understanding the pharmacokinetics and combining active targeting with passive targeting to further improve the therapeutic efficacy and increase survival.


Cancer Prevention Research | 2011

Phase 0 Clinical Chemoprevention Trial of the Akt Inhibitor SR13668

Joel M. Reid; Chad A. Walden; Rui Qin; Katie L. Allen Ziegler; John L. Haslam; Roger A. Rajewski; Roger A. Warndahl; Cindy L. Fitting; Daniel Boring; Eva Szabo; James A. Crowell; Marjorie Perloff; Ling Jong; Brent A. Bauer; Sumithra J. Mandrekar; Paul J. Limburg

SR13668, an orally active Akt pathway inhibitor, has demonstrated cancer chemopreventive potential in preclinical studies. To accelerate the clinical development of this promising agent, we designed and conducted the first-ever phase 0 chemoprevention trial to evaluate and compare the effects of food and formulation on SR13668 bioavailability. Healthy adult volunteers were randomly assigned to receive a single, 38-mg oral dose of SR13668 in one of five different formulations, with or without food. On the basis of existing animal data, SR13668 in a PEG400/Labrasol oral solution was defined as the reference formulation. Blood samples were obtained pre- and post-agent administration for pharmacokinetic analyses. Area under the plasma concentration–time curve (AUC0–∞) was defined as the primary endpoint. Data were analyzed and compared using established statistical methods for phase 0 trials with a limited sample size. Participants (n = 20) were rapidly accrued over a 5-month period. Complete pharmacokinetic data were available for 18 randomized participants. AUC0–∞ values were highest in the fed state (range = 122–439 ng/mL × hours) and were statistically significantly different across formulations (P = 0.007), with Solutol HS15 providing the highest bioavailability. SR13668 time to peak plasma concentration (3 hours; range, 2–6 hours) and half-life were (11.2 ± 3.1 hours) were not formulation-dependent. Using a novel, highly efficient study design, we rapidly identified a lead formulation of SR13668 for further clinical testing. Our findings support application of the phase 0 trial paradigm to accelerate chemoprevention agent development. Cancer Prev Res; 4(3); 347–53. ©2011 AACR.


Scientific Reports | 2015

Tuning Pharmacokinetics and Biodistribution of a Targeted Drug Delivery System Through Incorporation of a Passive Targeting Component

Rachel A. Kudgus; Chad A. Walden; Renee M. McGovern; Joel M. Reid; J. David Robertson; Priyabrata Mukherjee

Major challenges in the development of drug delivery systems (DDSs) have been the short half-life, poor bioavailability, insufficient accumulation and penetration of the DDSs into the tumor tissue. Understanding the pharmacokinetic (PK) parameters of the DDS is essential to overcome these challenges. Herein we investigate how surface chemistry affects the PK profile and organ distribution of a gold nanoparticle-based DDS containing both a passive and active targeting moiety via two common routes of administration: intravenous and intraperitoneal injections. Using LC/MS/MS, ELISA and INAA we report the half-life, peak plasma concentrations, area under the curve, ability to cross the peritoneal barrier and biodistribution of the nanoconjugates. The results highlight the design criteria for fine-tuning the PK parameters of a targeted drug delivery system that exploits the benefits of both active and passive targeting.


Journal of Drug Metabolism and Toxicology | 2016

Comparative Metabolism of Batracylin (NSC 320846) and N-acetylbatracylin (NSC 611001) Using Human, Dog, and Rat Preparations In Vitro

Joseph M. Covey; Joel M. Reid; Sarah A. Buhrow; Mary J. Kuffel; Chad A. Walden; Holger P. Behrsing

BACKGROUND Batracylin is a heterocyclic arylamine topoisomerase inhibitor with preclinical anticancer activity. Marked species differences in sensitivity to the toxicity of batracylin were observed and attributed to differential formation of N-acetylbatracylin by N-acetyltransferase. A Phase I trial of batracylin in cancer patients with slow acetylator genotypes identified a dose-limiting toxicity of hemorrhagic cystitis. To further explore the metabolism of batracylin and N-acetylbatracylin across species, detailed studies using human, rat, and dog liver microsomal and hepatocyte preparations were conducted. METHODS Batracylin or N-acetylbatracylin was incubated with microsomes and hepatocytes from human, rat, and dog liver and with CYP-expressing human and rat microsomes. Substrates and metabolites were analyzed by HPLC with diode array, fluorescence, radiochemical, or mass spectrometric detection. Covalent binding of radiolabeled batracylin and N-acetylbatracylin to protein and DNA was measured in 3-methylcholanthrene-induced rat, human, and dog liver microsomes, and with recombinant human cytochromes P450. RESULTS In microsomal preparations, loss of batracylin was accompanied by formation of one hydroxylated metabolite in human liver microsomes and five hydroxylated metabolites in rat liver microsomes. Six mono- or di-hydroxy-N-acetylbatracylin metabolites were found in incubations of this compound with 3MC rat liver microsomes. Hydroxylation sites were identified for some of the metabolites using deuterated substrates. Incubation with recombinant cytochromes P450 identified rCYP1A1, rCYP1A2, hCYP1A1 and hCYP1B1 as the major CYP isoforms that metabolize batracylin and N-acetylbatracylin. Glucuronide conjugates of batracylin were also identified in hepatocyte incubations. NADPH-dependent covalent binding to protein and DNA was detected in all batracylin and most N-acetylbatracylin preparations evaluated. CONCLUSIONS Microsomal metabolism of batracylin and N-acetylbatracylin results in multiple hydroxylated products (including possible hydroxylamines) and glutathione conjugates. Incubation of batracylin with hepatocytes resulted in production primarily of glucuronides and other conjugates. There was no clear distinction in the metabolism of batracylin and N-acetylbatracylin across species that would explain the differential toxicity.


Toxicologic Pathology | 2015

Characterization of Batracylin-induced Renal and Bladder Toxicity in Rats:

Myrtle A. Davis; Deborah I. Bunin; Steven Samuelsson; Kenneth P. Altera; Robert J. Kinders; Scott M. Lawrence; Jiuping Ji; Sarah A. Buhrow; Chad A. Walden; Joel M. Reid; Linda L. Rausch; Toufan Parman

Batracylin (NSC-320846) is a dual inhibitor of DNA topoisomerases I and II. Batracylin advanced as an anticancer agent to Phase I clinical trials where dose limiting hemorrhagic cystitis (bladder inflammation and bleeding) was observed. To further investigate batracylin’s mechanism of toxicity, studies were conducted in Fischer 344 rats. Once daily oral administration of 16 or 32 mg/kg batracylin to rats for 4 days caused overt toxicity. Abnormal clinical observations and adverse effects on clinical pathology, urinalysis, and histology indicated acute renal damage and urothelial damage and bone marrow dysfunction. Scanning electron microscopy revealed sloughing of the superficial and intermediate urothelial layers. DNA damage was evident in kidney and bone marrow as indicated by histone γ-H2AX immunofluorescence. After a single oral administration of 16 or 32 mg/kg, the majority of batracylin was converted to N-acetylbatracylin (NAB) with a half-life of 4 hr to 11 hr. Mesna (Mesnex™), a drug known to reduce the incidence of hemorrhagic cystitis induced by ifosfamide or cyclophosphamide, was administered to rats prior to batracylin, but did not alleviate batracylin-induced bladder and renal toxicity. These findings suggest that batracylin results in DNA damage-based mechanisms of toxicity and not an acrolein-based mechanism of toxicity as occurs after ifosfamide or cyclophosphamide administration.


Molecular Cancer Therapeutics | 2015

Abstract B113: Phase I and pharmacokinetic study of ixabepilone and temsirolimus in adult patients with advanced solid tumors

Joel M. Reid; Michael E. Menefee; Felix Boakye-Agyeman; Chad A. Walden; Charles Erlichman; Keith C. Bible

Background: The combination of a microtubule stabilizing agent and an mTOR inhibitor has been identified as a synergistic combination in preclinical models. Additional studies have further evaluated the impact of treatment sequence with paclitaxel and rapamycin where administration of rapamycin after paclitaxel was associated with synergy. In our preclinical studies, apoptosis was greater with the combination of ixabepilone and temsirolimus than with either single agent. Thus, a Phase I study of the combination of Ixabepilone (IXB) and Temsirolimus (TEM) was performed to determine the maximum tolerated dose (MTD), describe the toxicity profile and characterize the pharmacokinetics of each agent in patients with advanced cancer. Methods: Eligible patients included adults with a histologically confirmed solid tumor malignancy that was metastatic or unresectable who have received ≤ 2 chemotherapy regimens and had ECOG PS ≤ 2 with adequate bone marrow, renal and hepatic function. Using a standard 3+3 design, patients were treated with IXB IV over 3 hours on day 1 and TEM IV over 0.5 hours on days 2 and 9 (schedule A) or days 1 and 8 (schedule B) every 21 days. Pharmacokinetics (PK) was performed in patients on schedule B. Results: 22 evaluable patients were enrolled between August 24, 2011 and November 7, 2014. The final 7 patients were treated on schedule B. Patients were treated at 3 dose levels of IXB (mg/m2)/TEM (mg): DL1- 24/15 (A- 6 pts, B- 2 pts); DL2- 32/15 (A- 3 pts, B- 5 pts); DL3- 32/20 (A- 6 pts). One patient had DLT (grade 3 hypophosphatemia) and 1 patient expired due to disease progression in DL1A. No DLTs were seen in DL2A. The dose was escalated to DL3 and 1 patient had DLT (grade 3 hypokalemia). Following a change to schedule B, 5 patients were enrolled to DL2B and 1 experienced DLT (grade 4 neutropenia, grade 4 thrombocytopenia, grade 4 sepsis). The dose was reduced to DL1B and 2 more patients were enrolled, both of whom experienced DLTs (grade 5 bronchopulmonary hemorrhage and grade 4 neutropenia). Grade 4 events considered at least possibly related to treatment occurred in 6 of 22 patients (27%) and included cardiac arrest, dyspnea, hypokalemia, hypoxia, multi-organ failure, decreased platelets, acute renal failure, neutropenia, neutropenic fever, sepsis, and decreased WBCs. Patients received a median of 4 cycles (range, 1-10+). Partial response was noted in 1 patient and stable disease was noted in 11 patients. One patient remains on treatment after 11 cycles. IXB (plasma), TEM (whole blood) and sirolimus (whole blood) concentrations were measured by a sensitive, specific lc/ms/ms assay. A 24 and 32 mg/m2 IXB dose yields Cmax of 159 and 312 ng/ml, respectively. The IXB CLp and t1/2 values were 31 L/h and 76 h, respectively. The Cmax, Cl and t1/2 values after a 15 mg TEM dose were 518 ng/ml, 6.4 L/h and 11 h, respectively. The Cmax and t1/2 values of sirolimus after a 15 mg TEM dose were 37.6 ng/ml and 49 h, respectively. The PK of IXB and TEM in combination were similar to those reported for each drug alone. Conclusions: The reasons for the unpredictable, severe toxicity of the IXB/TEM combination are unclear. The MTD could not be determined, and enrollment was discontinued. This trial exemplifies challenges associated with development of drug combinations with mTOR inhibitors. Supported in part by UM1 CA186686 and P30 CA15083. Clinical trial information: NCT01375829 Citation Format: Joel M. Reid, Michael E. Menefee, Felix Boakye-Agyeman, Chad A. Walden, Charles Erlichman, Keith C. Bible. Phase I and pharmacokinetic study of ixabepilone and temsirolimus in adult patients with advanced solid tumors. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr B113.


Cancer Research | 2013

Abstract 4557: LC/MS/MS assay and mouse pharmacokinetics of the phenylurea thiocarbamate NSC 161128.

Chad A. Walden; Joel M. Reid; Renee M. McGovern; Joseh M. Covey

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC NSC 161128 is a phenylurea thiocarbamate that has shown differential activity in the NCI60 anticancer screen and in xenograft models. The best in vivo activity was observed against the DU-145 prostate cell line. Previous studies conducted by DTP/NCI demonstrated that NSC 161128 is rapidly metabolized after administration to mice or rats, with the major detected metabolite being N-methyl-N’-phenylurea (M10). The other putative metabolite(s) arising from cleavage of the N-S bond, dimethyldithiocarbamate (M85) and/or its disulfide [bis(dimethyldithiocarbamoyl)disulfide, M17] were not detected in animal plasma using available methodologies. NSC 161128 was also shown to be rapidly degraded in aqueous solution at low and high pH, forming M10 and M17. As part of the preclinical evaluation of NSC 161128, we developed a sensitive, specific LC/MS/MS method to quantitate NSC 161128, M10 and M17 in biological samples, explored the stability of NSC 161128 in cell culture medium, and characterized the pharmacokinetics and in vivo metabolism of NSC 161128 following i.p. administration to athymic nude mice. We developed and validated suitable positive ionization lc/ms/ms methodology for measuring NSC 161128, M10 and M17. The assay utilized liquid extraction with isopropanol:1-chlorobutane, 1:9 (v/v) and MS/MS detection with the 270>177, 151>94 and 241>120 transitions for NSC 161128, M10 and M17, respectively. Baseline separation was achieved on an Atlantis T3 column under a mobile phase consisting of 40:60 methanol:water containing 0.1% formic acid. Standard curves were linear over the concentration range 1.0-1000 ng/mL with a lower limit of detection of 1.0 ng/mL. The parent compound was stable under acidic and neutral conditions, but some degradation ( 40%) was noted within 30 minutes, and samples for pharmacokinetic studies were kept cold and processed quickly to prevent degradation. NSC 161128 pharmacokinetics were characterized in male CD1 mice after i.p. administration of a 200 mg/kg dose. The peak plasma concentration of 255 ng/ml was observed 5 minutes after drug administration and the plasma half-life was 138 min. The peak concentration of 14.7 μg/ml M10 was detected 15 min after the NSC 161128 dose and the half-life was 560 min. Analysis of the pharmacokinetics of the thiol metabolites will be presented. Supported by NCI contract N01-CM-2011-00014. Citation Format: Chad A. Walden, Joel M. Reid, Renee M. McGovern, Joseh M. Covey, Matthew M. Ames. LC/MS/MS assay and mouse pharmacokinetics of the phenylurea thiocarbamate NSC 161128. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4557. doi:10.1158/1538-7445.AM2013-4557


Cancer Research | 2012

Abstract 3769: Metabolic activation of batracylin and N-acetylbatracylin to reactive metabolites that bind protein and DNA: Possible role in toxicity

Sarah A. Buhrow; Mary J. Kuffel; Chad A. Walden; Joel M. Reid; Joseph M. Covey

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Batracylin (NSC 320846, BAT) is a heterocyclic arylamine and topoisomerase II inhibitor shown to be active in murine tumor models, but with demonstrated species differences in toxicity associated with metabolism to N-acetylbatracylin (NBAT) by N-acetyltransferase (NAT). BAT is a preferential substrate of human NAT2. Accordingly, production of NBAT should be subject to population variability as a function of NAT2 polymorphisms. A phase I clinical trial of BAT in patients with advanced cancer and slow acetylator NAT2 phenotypes to reduce the risk of NBAT-related toxicity found a dose-limiting toxicity (DLT) of cystitis/hemorrhagic cystitis. In subsequent in vitro and in vivo preclinical studies to elucidate metabolic pathways that might contribute to BAT toxicity, we found oxidative metabolites and thiol conjugates of BAT and NBAT (MM Ames et al, Cancer Res 2011;71(8 Suppl):Abstract nr 1303). Since these data are consistent with metabolic activation of BAT to potentially toxic metabolites, we assessed formation of reactive intermediates as measured by covalent binding (CB) to protein and DNA or by formation of glutathione conjugates. Radiolabeled BAT and NBAT were incubated with 3-MC-induced rat (r), human (h) and dog (d) liver microsomes (LM), as well as with recombinant human cytochromes P450 (CYPs) with or without added DNA. NADPH-dependent BAT protein and DNA CB were detected in all microsomal preparations (see table below). NADPH-dependent NBAT protein CB was detected in all microsomal preparations except hCYP1B1. NADPH-dependent NBAT DNA CB was detected in all microsomal preparations except 3MC-rLM. We also detected a glutathione conjugate of mono-hydroxy BAT by lc/ms/ms analysis when BAT was incubated with hLM in the presence of glutathione and NADPH. Thus, CYP-catalyzed oxidation of BAT yields reactive metabolites that that bind protein and DNA and may contribute to drug-related DLT. Supported in part by NCI Contract N01-CM-52206. ![Figure][1] Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3769. doi:1538-7445.AM2012-3769 [1]: pending:yes


Cancer Research | 2012

Abstract 3781: Pharmacokinetics of endoxifen and tamoxifen in female mice: Implications for comparative in vivo activity studies

Stephanie L. Safgren; Sarah A. Buhrow; Chad A. Walden; Mary J. Kuffel; Kathryn E. Reinicke; Joel M. Reid; Matthew P. Goetz

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Background: In tamoxifen (TAM) treated patients, both CYP2D6 metabolism and low steady state concentrations of endoxifen (END), the active metabolite of TAM, have been associated with a higher risk of recurrence (Madlensky L, Clin Pharmacol Ther. 2011; 89(5):718-25). END drug development is ongoing at Mayo and NCI, and our previous data demonstrated that 1) END had high oral bioavailability in mice and yielded plasma concentrations and total exposure 20-fold greater as compared to an equivalent oral dose of TAM (MM Ames et al, Cancer Res 2011;70(8 Suppl 1):Abstract nr 3603) and 2) END is superior to TAM in both HER2 (Reinicke AACR 2011) and aromatase expressing (Goetz, SABC 2011) MCF-7 xenograft models. While subcutaneous (SQ) TAM is used by most researchers to study and develop models of TAM resistance, little is known about 1) whether murine and human TAM pharmacokinetics are comparable and 2) whether the route of administration affects concentrations of the active metabolites. Therefore, we conducted murine pharmacokinetic studies evaluating both the oral and SQ administration of TAM and END. Results: Following administration of TAM, concentrations of the active metabolites (END and 4-hydroxy TAM [4HT]) were therapeutic (Cmax- END 35.9 nM, 4HT 62.8 nM) only after a 0.5 mg oral dose of TAM. In contrast, following all doses of SQ TAM, concentrations of the active metabolites (END and 4HT) were sub-therapeutic (Cmax- END 5.2 nM; 4HT 18.7 nM). In contrast to humans, the Cmax of N-desmethyl TAM, the most abundant human TAM metabolite, was 80 nM) were detected in plasma at every dose and both dosing routes. Comparable END and TAM plasma concentrations were achieved with a 0.0625 mg SQ END and a 0.5 mg SQ TAM, respectively. Conclusions: Our data strongly suggest that SQ dosing of TAM results in non-therapeutic concentrations of the active hydroxylated metabolites, and may be a simple pharmacokinetic factor influencing response and resistance in a murine in vivo xenograft model. Given the nonlinear pharmacokinetics and differences in metabolism, the dose and route of END must be carefully selected for comparative studies with TAM. These data support the ongoing human studies designed to bypass the need for TAM metabolism by the direct administration of END. Supported by the Mayo Comprehensive Cancer Center Grant (CA15083) and the Mayo Clinic Breast Cancer SPORE (CA 116201; MMA, JMR, MPG). Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3781. doi:1538-7445.AM2012-3781

Collaboration


Dive into the Chad A. Walden's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph M. Covey

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Priyabrata Mukherjee

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel Boring

New York Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge