Network


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

Hotspot


Dive into the research topics where Michael Baltezor is active.

Publication


Featured researches published by Michael Baltezor.


Oncotarget | 2017

In silico and in vitro drug screening identifies new therapeutic approaches for Ewing sarcoma

Ziyan Y. Pessetto; Bin Chen; Hani J Alturkmani; Stephen Hyter; Colleen A. Flynn; Michael Baltezor; Yan Ma; Howard G. Rosenthal; Kathleen Neville; Scott Weir; Atul J. Butte; Andrew K. Godwin

The long-term overall survival of Ewing sarcoma (EWS) patients remains poor; less than 30% of patients with metastatic or recurrent disease survive despite aggressive combinations of chemotherapy, radiation and surgery. To identify new therapeutic options, we employed a multi-pronged approach using in silico predictions of drug activity via an integrated bioinformatics approach in parallel with an in vitro screen of FDA-approved drugs. Twenty-seven drugs and forty-six drugs were identified, respectively, to have anti-proliferative effects for EWS, including several classes of drugs in both screening approaches. Among these drugs, 30 were extensively validated as mono-therapeutic agents and 9 in 14 various combinations in vitro. Two drugs, auranofin, a thioredoxin reductase inhibitor, and ganetespib, an HSP90 inhibitor, were predicted to have anti-cancer activities in silico and were confirmed active across a panel of genetically diverse EWS cells. When given in combination, the survival rate in vivo was superior compared to auranofin or ganetespib alone. Importantly, extensive formulations, dose tolerance, and pharmacokinetics studies demonstrated that auranofin requires alternative delivery routes to achieve therapeutically effective levels of the gold compound. These combined screening approaches provide a rapid means to identify new treatment options for patients with a rare and often-fatal disease.


Veterinary and Comparative Oncology | 2018

Evaluation of intravenous and subcutaneous administration of a novel, excipient-free, nanoparticulate formulation of paclitaxel in dogs with spontaneously occurring neoplasia

Kim A. Selting; Sandra M. Bechtel; Jahna Espinosa; Carolyn J. Henry; Deborah Tate; Jeffrey N. Bryan; Lian G. Rajewski; Brian K. Flesner; Charles J. Decedue; Michael Baltezor

Carriers used to solubilize taxane chemotherapy drugs cause severe hypersensitivity. Nanoparticle formulations can provide improved dissolution and bioavailability of taxanes. Thus, a nanoparticulate, excipient-free formulation of paclitaxel (CTI52010) was evaluated in tumour-bearing dogs with intravenous and subcutaneous delivery. Tumour-bearing dogs were treated with intravenous CTI52010 using a modified rapid dose escalation scheme. Subcutaneous administration was then planned for a small cohort of dogs for comparison. For both groups, serial blood samples were collected after first dosing for pharmacokinetic analysis by LCMSMS. Tumour response was measured using RECIST criteria. Toxicity was recorded using VCOG-CTCAEv1.1. Fifteen dogs were treated with intravenous delivery at increasing dosages (80-136 mg/m2 ), with one objective response in the urethral component of a prostatic carcinoma (probable transitional cell carcinoma) and four dogs with durable stable disease (two carcinomas, two sarcomas). Pharmacokinetic data indicate a rapid initial clearing of the drug from serum followed by an extended elimination half-life, similar to normal dogs and suggesting reticuloendothelial clearance. Parameters and toxicity were highly variable and a maximally tolerated dosage could not be reliably confirmed. Three dogs were treated with subcutaneous delivery and no drug was detected in circulation, resulting in termination of the study. This novel formulation of paclitaxel is well tolerated in dogs and no unique toxicity or hypersensitivity was noted. The preliminary responses suggest biologic activity. The lack of systemic absorption after subcutaneous administration suggests a possible role for intratumoural anticancer therapy.


Journal of Aerosol Medicine and Pulmonary Drug Delivery | 2018

Pharmacokinetic Profile of Inhaled Submicron Particle Paclitaxel (NanoPac®) in a Rodent Model

James Verco; William Johnston; Michael Baltezor; Philip J. Kuehl; Andrew P. Gigliotti; Steven A. Belinsky; Anita Lopez; Ronald Wolff; Lauren Hylle; Gere S. diZerega

Abstract Background: Inhaled chemotherapeutics may enhance pulmonary drug exposure to malignant lesions in the lung without substantially contributing to systemic toxicities. The pharmacokinetic profile of inhaled submicron particle paclitaxel (NanoPac®) in healthy rodent plasma and lung tissue is evaluated here to determine administration proof-of-principle. Methods: Healthy male Sprague Dawley rats received paclitaxel in one of three arms: intravenous nab-paclitaxel at 2.9 mg/kg (IVnP), inhaled NanoPac low dose (IHNP-LD) at 0.38 mg/kg, or inhaled NanoPac high dose (IHNP-HD) at 1.18 mg/kg. Plasma and lung tissue paclitaxel concentrations were determined using ultraperformance liquid chromatography tandem mass spectrometry from animals sacrificed at 10 time points ranging up to 2 weeks after administration. Peak concentration (Cmax), apparent residence half-life (T1/2), exposure (AUC(last)), and dose-normalized exposure (AUCD(last)) were determined. Pulmonary histopathology was performed on rats sacrificed at the 336-hour time point. Results: Paclitaxel was detectable and quantifiable in the rat lung for both inhaled NanoPac arms sampled at the final necropsy, 336 hours postadministration. Substantial paclitaxel deposition and retention resulted in an order of magnitude increase in dose-normalized pulmonary exposure over IVnP. Inhaled NanoPac arms had an order of magnitude lower plasma Cmax than IVnP, but followed a similar plasma T1/2 clearance (quantifiable only to 72 hours postadministration). Pulmonary histopathology found all treated animals indistinguishable from treatment-naive rats. Conclusion: In the rodent model, inhaled NanoPac demonstrated substantial deposition and retention of paclitaxel in sampled lung tissue. Further research to determine NanoPacs toxicity profile and potential efficacy as lung cancer therapy is underway.


Cancer Research | 2018

Abstract 5882: Bench-to-bedside translation of ciclopirox prodrug for the treatment of non-muscle invasive and muscle-invasive bladder cancer

Scott Weir; Partha Ranjarajan; Robyn Wood; Karl Schorno; Prabhu Ramamoorthy; Lian Rajweski; Kathy Heppert; Michael J. McKenna; William McCulloch; Greg Reed; Amanda E. Brinker; Michael Baltezor; Roy A. Jensen; John A. Taylor; Shrikant Anant

Ciclopirox (CPX) is contained in a number of FDA-approved topical antifungal drug products as the free acid and olamine salt. CPX possesses anticancer activity in a number of in vitro and in vivo preclinical models. Its clinical utility is limited as an oral anticancer agent, however. The oral bioavailability of CPX is quite low due to extensive first pass effect. The poor water solubility of CPX and its olamine salt prevent formulation as an injectable drug product. Thirdly, dose-limiting gastrointestinal toxicities were observed following four times daily oral dosing of CPX in patients with advanced hematologic malignancies. Ciclopirox Prodrug (CPX-POM), in contrast, has demonstrated excellent bioavailability via injectable routes of administration. Here we describe the preclinical characterization of CPX-POM, a novel anticancer agent being developed for the treatment of non-muscle invasive (NMIBC) and muscle invasive (MIBC) bladder cancer. Following IV, SQ and IP administration to mice, CPX-POM is rapidly and completely metabolized to CPX in blood via circulating phosphatases. CPX and its major, inactive glucuronide metabolite are extensively eliminated in urine. At well-tolerated doses, steady-state urine concentrations of CPX exceed in vitro IC50 values in mice by 15-30 fold. CPX inhibited cell proliferation, colony formation, and bladdosphere formation in vitro in T24 (NMIBC) and 253JBV (MIBC) human cell lines in both concentration- and time-dependent manners with IC50 values of 2-4 µM. CPX exposure increased the percentage of NMIBC and MIBC cells arrested at the S and G0/G1 phases, and induced cell death. CPX exposure significantly reduced expression of genes at the mRNA level involved in cancer stem cell signaling pathways including Notch, Wnt, and Hedgehog. CPX was shown to inhibit bladder cancer cell growth in vitro by inhibiting the Notch 1 signaling pathway. The validated N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN) chemical carcinogen mouse model of bladder cancer was employed to establish in vivo preclinical proof of principle for CPX-POM. Over the once-daily IP dose range of 25-200 mg/kg, CPX-POM treatment resulted in significant decreases in bladder weight, a clear migration to lower stage tumors, dose-dependent reduction in Ki67 and PCNA staining, as well as a reduction in PCNA-expressing cells. All CPX-POM doses were well tolerated with no evidence of toxicity to the urinary tract based on blinded pathologic evaluation. There were also dose-dependent decreases in Notch 1, Presenilin 1, and Hey 1 in bladder cancer tissues obtained from CPX-POM treated animals. Tumor response was similar, in vivo, following once-daily and three-times weekly CPX-POM administration. CPX-POM has received FDA clearance to proceed to Phase I, and is currently being evaluated in a first-in-human trial in patients with advanced solid tumors. Citation Format: Scott J. Weir, Partha Ranjarajan, Robyn Wood, Karl Schorno, Prabhu Ramamoorthy, Lian Rajweski, Kathy Heppert, Michael J. McKenna, William McCulloch, Greg A. Reed, Amanda Brinker, Michael J. Baltezor, Roy A. Jensen, John A. Taylor, Shrikant Anant. Bench-to-bedside translation of ciclopirox prodrug for the treatment of non-muscle invasive and muscle-invasive bladder cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5882.


ACS Chemical Neuroscience | 2018

Tyr1-ψ[(Z)CF═CH]-Gly2 Fluorinated Peptidomimetic Improves Distribution and Metabolism Properties of Leu-Enkephalin

Ryan A. Altman; Krishna Sharma; Lian G. Rajewski; Paul Chester Toren; Michael Baltezor; Mohan Pal; Somnath Narayan Karad

Opioid peptides are key regulators in cellular and intercellular physiological responses, and could be therapeutically useful for modulating several pathological conditions. Unfortunately, the use of peptide-based agonists to target centrally located opioid receptors is limited by poor physicochemical (PC), distribution, metabolic, and pharmacokinetic (DMPK) properties that restrict penetration across the blood-brain barrier via passive diffusion. To address these problems, the present paper exploits fluorinated peptidomimetics to simultaneously modify PC and DMPK properties, thus facilitating entry into the central nervous system. As an initial example, the present paper exploited the Tyr1-ψ[( Z)CF═CH]-Gly2 peptidomimetic to improve PC druglike characteristics (computational), plasma and microsomal degradation, and systemic and CNS distribution of Leu-enkephalin (Tyr-Gly-Gly-Phe-Leu). Thus, the fluoroalkene replacement transformed an instable in vitro tool compound into a stable and centrally distributed in vivo probe. In contrast, the Tyr1-ψ[CF3CH2-NH]-Gly2 peptidomimetic decreased stability by accelerating proteolysis at the Gly3-Phe4 position.


Cancer Research | 2015

Abstract 1895: Ciclopirox prodrug for the prevention and therapy of non-muscle invasive bladder cancer

Parthasarathy Rangarajan; Dharmalingam Subramaniam; Michael Baltezor; Robyn Wood; Shrikant Anant; Scott Weir

Objective: Ciclopirox prodrug for the prevention of Non-Muscle Invasive Bladder Cancer (NMIBC) represents a potential breakthrough in the management of high-grade, NMIBC. If successful, this represents the first systemic approach to manage NMIBC. Ciclopirox olamine, the active pharmaceutical ingredient in several topical antifungal drug products, has demonstrated anticancer activity in blood and solid tumors. We developed a phosphoryloxymethyl prodrug of ciclopirox (CPX) for efficient intravenous delivery, thereby avoiding dose-limiting gastrointestinal toxicities and first-pass effect associated with oral administration. Methods: In vitro anticancer activity of CPX was performed in both NMIBC (T24) and MIBC (253-JBV) cell lines. Cell proliferation was determined by hexoseaminidase assay. Stemness was determined by the Spheroid assay. Flow cytometric analyses was performed for cell cycle analysis. RT-PCR array analysis was performed to identify signaling pathways. Real Time PCR, western blot and immunofluorescent studies were performed for determining gene expression. Results: Pharmacokinetic studies conducted in mice, rats and dogs demonstrated that the drug is rapidly and completely metabolized and eliminated via the urine. In vitro, CPX inhibited growth of T24 and 253-JBV cells at 4 and 2 μM, respectively coupled with S-phase cell cycle arrest. FITC conjugated Annexin V-coupled flow cytometry studies showed that CPX induces apoptotic cell death. CPX also suppressed bladdosphere formation, suggesting that stem cells are affected. Notch signaling pathway plays a significant role in stem cell behavior, and previous studies have suggested that the pathway may be a therapeutic target for bladder cancer. Notch receptor activation, which occurs following ligand binding, involves specific intracellular cleavage by the γ-secretase complex. First, RT-PCR array analyses suggested that CPX inhibits the Notch signaling pathway. Furthermore, CPX significantly inhibited the expression of the γ-secretase complex proteins Presenilin1, Nicastrin, APH-1 and PEN-2. In addition, there was a reduction in the expression of downstream targets of the pathway including Hes1 and Cyclin-D1. These data suggest that CPX significantly inhibits Notch intracellular signaling pathway proteins. Ectopic expression of the activated/cleaved Notch protein reversed CPX mediated inhibition of cell proliferation. Conclusion: CPX is a novel chemotherapeutic and preventive agent for bladder cancers, and this occurs by suppressing the stem cells. We are confident that there will be no failure of the compound as a result of unacceptable toxicity because drug safety profile of CPX has been previously characterized in animals and humans. These studies provide insight into understanding the role of CPX as a potent chemotherapeutic and preventive agent. Citation Format: Parthasarathy Rangarajan, Satish Ramalingam, Dharmalingam Subramaniam, Michael J. Baltezor, Robyn Wood, Shrikant Anant, Scott Weir. Ciclopirox prodrug for the prevention and therapy of non-muscle invasive bladder cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1895. doi:10.1158/1538-7445.AM2015-1895


Cancer Chemotherapy and Pharmacology | 2015

A phase I study of intraperitoneal nanoparticulate paclitaxel (Nanotax®) in patients with peritoneal malignancies

Stephen K. Williamson; Gary Johnson; Holly Maulhardt; Kathleen M. Moore; D. S. McMeekin; Thomas K. Schulz; Gregory A. Reed; Katherine F. Roby; Christine B. Mackay; Holly J. Smith; Scott Weir; Jo Wick; Maurie Markman; Gere S. diZerega; Michael Baltezor; Jahna Espinosa; Charles J. Decedue


Archive | 2016

Early Drug Discovery and Development Guidelines: For Academic Researchers, Collaborators, and Start-up Companies

Jeffrey Strovel; Sitta Sittampalam; Nathan P. Coussens; Michael Hughes; James Inglese; Andrew Kurtz; Ali Andalibi; Lavonne Patton; Christopher M. Austin; Michael Baltezor; Michael Beckloff; Michael Weingarten; Scott Weir


Archive | 2015

Use of Paclitaxel Particles

Stephen K. Williamson; Charles J. Decedue; Steven A. Fontana; Michael Baltezor


Journal of Clinical Oncology | 2018

NanoPac inhalation treatment of NSCLC in a nude rat orthotopic lung cancer model.

Gere S. diZerega; William Johnston; James Verco; Michael Baltezor; Ronald Wolff; Philip Kuehl

Collaboration


Dive into the Michael Baltezor's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

James Inglese

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Nathan P. Coussens

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Christopher P. Austin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gere S. diZerega

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge