Network


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

Hotspot


Dive into the research topics where Andrew S. Holpuch is active.

Publication


Featured researches published by Andrew S. Holpuch.


Pharmaceutical Research | 2010

Nanoparticles for Local Drug Delivery to the Oral Mucosa: Proof of Principle Studies

Andrew S. Holpuch; Garrett J. Hummel; Meng Tong; Garrett A. Seghi; Ping Pei; Ping Ma; Russell J. Mumper; Susan R. Mallery

ABSTRACTPurposeTo determine if solid lipid nanoparticles represent a viable strategy for local delivery of poorly water soluble and unstable chemopreventive compounds to human oral tissues.MethodsNanoparticle uptake and compound retention evaluations employed monolayer-cultured human oral squamous cell carcinoma (OSCC) cell lines and normal human oral mucosal explants. Feasibility of nanoparticle delivery was also evaluated with respect to the presence of phase-III efflux transporters in normal oral mucosal tissue and OSCC tissues.ResultsFunctional uptake assays confirmed significantly greater internalization of nanoparticle-delivered fluorescent probe relative to free-fluorescent probe delivery, while concurrently demonstrating nanoparticle uptake rate differences among the OSCC cell lines and the phagocytic control human monocyte cell line. Mucosal explants exhibited nanoparticle penetration and internalization in the spinous and basal epithelial layers (7/10 specimens), and also exhibited the presence of the phase-III efflux transporters multidrug resistance-associated protein 1 (MRP1) and breast cancer resistance protein (BCRP).ConclusionsThese data confirm nanoparticle internalization by OSCC cells and support the premise that nanoparticle-based delivery provides higher final intracellular levels relative to bolus administration. Furthermore, the penetration and subsequent internalization of nanoparticles within the proliferating basal layer cells demonstrates the feasibility of nanoparticle formulations for local delivery and stabilization of oral chemopreventive compounds.


Cancer Prevention Research | 2011

Effects of human oral mucosal tissue, saliva and oral microflora on intraoral metabolism and bioactivation of black raspberry anthocyanins

Susan R. Mallery; Deric E. Budendorf; Matthew P. Larsen; Ping Pei; Meng Tong; Andrew S. Holpuch; Peter E. Larsen; Gary D. Stoner; Henry W. Fields; Kenneth K. Chan; Yonghua Ling; Zhongfa Liu

Our oral cancer chemoprevention trial data implied that patient-specific differences in local retention and metabolism of freeze-dried components of black raspberries (BRB) affected therapeutic responsiveness. Subsequent studies have confirmed that anthocyanins are key contributors to BRBs chemopreventive effects. Consequently, functional assays, immunoblotting, and immunohistochemical analyses to evaluate levels and distribution of BRB anthocyanin-relevant metabolic enzymes in human oral tissues were conducted. Liquid chromatography/tandem mass spectrometry (LC/MS-MS) analyses of time course saliva samples collected following BRB rinses were conducted to assess local pharmacokinetics and compare the capacities of three different BRB rinse formulations to provide sustained intraoral levels of anthocyanins. Protein profiles showed the presence of key metabolic enzymes in all 15 oral mucosal tissues evaluated, whereas immunohistochemistry confirmed these enzymes were distributed within surface oral epithelia and terminal salivary ducts. β-Glucosidase assays confirmed that whole and microflora-reduced saliva can deglycosylate BRB anthocyanins, enabling generation of the bioactive aglycone, cyanidin. LC/MS-MS analyses showed retention of parent anthocyanins and their functional, stable metabolite, protocatechuic acid, in saliva for up to 4 hours after rinsing. Furthermore, postrinse saliva samples contained glucuronidated anthocyanin conjugates, consistent with intracellular uptake and phase II conversion of BRB anthocyanins into forms amenable to local recycling. Our data show that comparable to the small intestine, the requisite hydrolytic, phase II and efflux transporting enzymes necessary for local enteric recycling are present and functional in human oral mucosa. Notably, interpatient differences in anthocyanin bioactivation and capacities for enteric recycling would impact treatment as retention of bioactivated chemopreventives at the target site would sustain therapeutic effectiveness. Cancer Prev Res; 4(8); 1209–21. ©2011 AACR.


Journal of Carcinogenesis | 2011

Optimizing therapeutic efficacy of chemopreventive agents: A critical review of delivery strategies in oral cancer chemoprevention clinical trials

Andrew S. Holpuch; Kashappa-Goud Desai; Steven P. Schwendeman; Susan R. Mallery

Due to its characterized progression from recognized premalignant oral epithelial changes (i.e., oral epithelial dysplasia) to invasive cancer, oral squamous cell carcinoma represents an optimal disease for chemopreventive intervention prior to malignant transformation. The primary goal of oral cancer chemoprevention is to reverse, suppress, or inhibit the progression of premalignant lesions to cancer. Over the last several decades, numerous oral cancer chemoprevention clinical trials have assessed the therapeutic efficacy of diverse chemopreventive agents. The standard of care for more advanced oral dysplastic lesions entails surgical excision and close clinical follow-up due to the potential (~33%) for local recurrence at a similar or more advanced histological stage. The purpose of this review was to identify prominent oral cancer chemoprevention clinical trials, assess their overall therapeutic efficacy, and delineate effects of local versus systemic drug administration. In addition, these compiled clinical trial data present concepts for consideration in the design and conduction of future clinical trials.


Molecular Pharmaceutics | 2012

Mucoadhesive fenretinide patches for site-specific chemoprevention of oral cancer: enhancement of oral mucosal permeation of fenretinide by coincorporation of propylene glycol and menthol.

Xiao Wu; Kashappa Goud H. Desai; Susan R. Mallery; Andrew S. Holpuch; Maynard P. Phelps; Steven P. Schwendeman

The objective of this study was to enhance oral mucosal permeation of fenretinide by coincorporation of propylene glycol (PG) and menthol in fenretinide/Eudragit RL PO mucoadhesive patches. Fenretinide is an extremely hydrophobic chemopreventive compound with poor tissue permeability. Coincorporation of 5-10 wt % PG (mean J(s) = 16-23 μg cm⁻² h⁻¹; 158-171 μg of fenretinide/g of tissue) or 1-10 wt % PG + 5 wt % menthol (mean J(s) = 18-40 μg cm⁻² h⁻¹; 172-241 μg of fenretinide/g of tissue) in fenretinide/Eudragit RL PO patches led to significant ex vivo fenretinide permeation enhancement (p < 0.001). Addition of PG above 2.5 wt % in the patch resulted in significant cellular swelling in the buccal mucosal tissues. These alterations were ameliorated by combining both enhancers and reducing PG level. After buccal administration of patches in rabbits, in vivo permeation of fenretinide across the oral mucosa was greater (∼43 μg fenretinide/g tissue) from patches that contained optimized permeation enhancer content (2.5 wt % PG + 5 wt % menthol) relative to permeation obtained from enhancer-free patch (∼17 μg fenretinide/g tissue) (p < 0.001). In vitro and in vivo release of fenretinide from patch was not significantly increased by coincorporation of permeation enhancers, indicating that mass transfer across the tissue, and not the patch, largely determined the permeation rate control in vivo. As a result of its improved permeation and its lack of deleterious local effects, the mucoadhesive fenretinide patch coincorporated with 2.5 wt % PG + 5 wt % menthol represents an important step in the further preclinical evaluation of oral site-specific chemoprevention strategies with fenretinide.


Carcinogenesis | 2012

Evaluation of a mucoadhesive fenretinide patch for local intraoral delivery: a strategy to reintroduce fenretinide for oral cancer chemoprevention

Andrew S. Holpuch; Maynard P. Phelps; Kashappa Goud H. Desai; Wei Chen; George Koutras; Byungdo B. Han; Blake M. Warner; Ping Pei; Garrett A. Seghi; Meng Tong; Michael B. Border; Henry W. Fields; Gary D. Stoner; Peter E. Larsen; Zhongfa Liu; Steven P. Schwendeman; Susan R. Mallery

Systemic delivery of fenretinide in oral cancer chemoprevention trials has been largely unsuccessful due to dose-limiting toxicities and subtherapeutic intraoral drug levels. Local drug delivery, however, provides site-specific therapeutically relevant levels while minimizing systemic exposure. These studies evaluated the pharmacokinetic and growth-modulatory parameters of fenretinide mucoadhesive patch application on rabbit buccal mucosa. Fenretinide and blank-control patches were placed on right/left buccal mucosa, respectively, in eight rabbits (30 min, q.d., 10 days). No clinical or histological deleterious effects occurred. LC-MS/MS analyses of post-treatment samples revealed a delivery gradient with highest fenretinide levels achieved at the patch-mucosal interface (no metabolites), pharmacologically active levels in fenretinide-treated oral mucosa (mean: 5.65 μM; trace amounts of 4-oxo-4-HPR) and undetectable sera levels. Epithelial markers for cell proliferation (Ki-67), terminal differentiation (transglutaminase 1-TGase1) and glucuronidation (UDP-glucuronosyltransferase1A1-UGT1A1) exhibited fenretinide concentration-specific relationships (elevated TGase1 and UGT1A1 levels <5 μM, reduced Ki-67 indices >5 μM) relative to blank-treated epithelium. All fenretinide-treated tissues showed significantly increased intraepithelial apoptosis (TUNEL) positivity, implying activation of intersecting apoptotic and differentiation pathways. Human oral mucosal correlative studies showed substantial interdonor variations in levels of the enzyme (cytochrome P450 3A4-CYP3A4) responsible for conversion of fenretinide to its highly active metabolite, 4-oxo-4-HPR. Complementary in vitro assays in human oral keratinocytes revealed fenretinide and 4-oxo-4-HPRs preferential suppression of DNA synthesis in dysplastic as opposed to normal oral keratinocytes. Collectively, these data showed that mucoadhesive patch-mediated fenretinide delivery is a viable strategy to reintroduce a compound known to induce keratinocyte differentiation to human oral cancer chemoprevention trials.


Experimental Cell Research | 2013

Inherent phenotypic plasticity facilitates progression of head and neck cancer: endotheliod characteristics enable angiogenesis and invasion.

Meng Tong; Byungdo B. Han; Andrew S. Holpuch; Ping Pei; Lingli He; Susan R. Mallery

The presence of the EMT (epithelial-mesenchymal transition), EndMT (endothelial-mesenchymal transition) and VM (vasculogenic mimicry) demonstrates the multidirectional extent of phenotypic plasticity in cancers. Previous findings demonstrating the crosstalk between head and neck squamous cell carcinoma (HNSCC) and vascular endothelial growth factor (VEGF) imply that HNSCC cells share some functional commonalities with endothelial cells. Our current results reveal that cultured HNSCC cells not only possess endothelial-specific markers, but also display endotheliod functional features including low density lipoprotein uptake, formation of tube-like structures on Matrigel and growth state responsiveness to VEGF and endostatin. HNSCC cell subpopulations are also highly responsive to transforming growth factor-β1 and express its auxiliary receptor, endoglin. Furthermore, the endotheliod characteristics observed in vitro recapitulate phenotypic features observed in human HNSCC tumors. Conversely, cultured normal human oral keratinocytes and intact or ulcerated human oral epithelia do not express comparable endotheliod characteristics, which imply that assumption of endotheliod features is restricted to transformed keratinocytes. In addition, this phenotypic state reciprocity facilitates HNSCC progression by increasing production of factors that are concurrently pro-proliferative and pro-angiogenic, conserving cell energy stores by LDL internalization and enhancing cell mobility. Finally, recognition of this endotheliod phenotypic transition provides a solid rationale to evaluate the antitumorigenic potential of therapeutic agents formerly regarded as exclusively angiostatic in scope.


Cancer Prevention Research | 2015

Fenretinide Perturbs Focal Adhesion Kinase in Premalignant and Malignant Human Oral Keratinocytes. Fenretinide's chemopreventive mechanisms include ECM interactions.

Byungdo B. Han; Suyang Li; Meng Tong; Andrew S. Holpuch; Richard Spinney; Daren Wang; Michael B. Border; Zhongfa Liu; Sachin C. Sarode; Ping Pei; Steven P. Schwendeman; Susan R. Mallery

The membrane-associated protein, focal adhesion kinase (FAK), modulates cell–extracellular matrix interactions and also conveys prosurvival and proliferative signals. Notably, increased intraepithelial FAK levels accompany transformation of premalignant oral intraepithelial neoplasia (OIN) to oral squamous cell carcinoma (OSCC). OIN chemoprevention is a patient-centric, optimal strategy to prevent OSCCs comorbidities and mortality. The cancer chemopreventive and synthetic vitamin A derivative, fenretinide, has demonstrated protein-binding capacities, for example, mTOR- and retinol-binding protein interactions. These studies used a continuum of human oral keratinocytes (normal-HPV E6/E7-transduced-OSCC) to assess potential fenretinide–FAK drug protein interactions and functional consequences on cellular growth regulation and motility. Molecular modeling studies demonstrated that fenretinide has approximately 200-fold greater binding affinity relative to the natural ligand (ATP) at FAKs kinase domain. Fenretinide also shows intermediate binding at FAKs FERM domain and interacts at the ATP-binding site of the closest FAK analogue, PYK2. Fenretinide significantly suppressed proliferation via induction of apoptosis and G2–M cell-cycle blockade. Fenretinide-treated cells also demonstrated F-actin disruption, significant inhibition of both directed migration and invasion of a synthetic basement membrane, and decreased phosphorylation of growth-promoting kinases. A commercially available FAK inhibitor did not suppress cell invasion. Notably, although FAKs FERM domain directs cell invasion, FAK inhibitors target the kinase domain. In addition, FAK-specific siRNA–treated cells showed an intermediate cell migration capacity; data which suggest cocontribution of the established migrating-enhancing PYK2. Our data imply that fenretinide is uniquely capable of disrupting FAKs and PYK2′s prosurvival and mobility-enhancing effects and further extend fenretinides chemopreventive contributions beyond induction of apoptosis and differentiation. Cancer Prev Res; 8(5); 419–30. ©2015 AACR.


Archive | 2016

Assessing the Changing Oral and Pharyngeal Cancer Demographic in the United States

Andrew S. Holpuch; Susan R. Mallery

In 2015, oral and pharyngeal cancer (OPC) will be diagnosed in an estimated 45,780 people in the United States (71.3 % male – accounting for 4 % of all cancer diagnoses in men) and 8650 deaths (69.4 % male) will be attributed to this disease. While the 5-year survival rate has slowly improved over the last several decades, mirroring an overall decline in tobacco use, the incidence has increased largely due to a rise in human papillomavirus (HPV)-associated oral and pharyngeal cancers. These primary risk factors (i.e., tobacco use and HPV infection), along with alcohol consumption, dietary patterns, immunosuppression, and genetic predisposition, are introduced relative to their role in the development of OPC. Concepts for the detection (clinical tools and appearance of precancerous lesions) and prevention (behavioral modification and HPV vaccination) of OPC are also presented.


Cancer Research | 2011

Abstract 3375: The epithelial to endotheliod transition (EET): A cellular modulation that promotes the head and neck squamous cell carcinoma tumorigenic phenotype

Meng Tong; Lingli He; Byungdo B. Han; Andrew S. Holpuch; Ping Pei; Susan R. Mallery

The presence of the EMT (epithelial-mesenchymal transition) and EndMT (endothelial-mesenchymal transition) demonstrates the extent of phenotypic plasticity in cancers. Previous findings in head and neck squamous cell carcinoma (HNSCC) cells, i.e., VEGF9s autocrine-paracrine function and their responsiveness to endostatin, imply that HNSCC cells share some functional characteristics with endothelial cells and suggest the potential for an “epithelial-to-endotheliod transition (EET)”. Such a transition would facilitate HNSCC tumorigenesis by augmenting release of proangiogenic factors and facilitating tumor cell migration and invasion. Consequently, these studies elucidated whether or not an EET occurs in HNSCC cells and tumors, and evaluated cellular parameters to uniquely characterize this epithelial-endotheliod transition. Our results demonstrate that cultured HNSCC cells express a variety of endothelial-specific markers, and assumption of the endotheliod phenotype has functional consequences including uptake of acetylated low density lipoprotein and enhanced invasion and migration. Selective HNSCC cells were also highly responsive to transforming growth factor-β1 and express its auxiliary receptor, endoglin. Furthermore, the EET is not restricted to cultured HNSCC cells per se as tumor cells in resected human HNSCC tumors and proliferating epithelia at the margins of ulcers demonstrate a similar phenotype. These data suggest that, similar to EMT, the EET enhances epithelial cell migration. Also, the regulation of this transition-transient (wound healing) versus sustained (invasive cancers)-impacts clinical outcome. Finally, recognition of the EET as a distinct entity provides impetus for future clinical trials to concurrently assess the antitumorigenic as well as the angiostatic properties of therapeutic agents formally regarded as exclusively angiostatic in scope. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3375. doi:10.1158/1538-7445.AM2011-3375


Pharmaceutical Research | 2011

Development and in vitro-in vivo evaluation of fenretinide-loaded oral mucoadhesive patches for site-specific chemoprevention of oral cancer.

Kashappa Goud H. Desai; Susan R. Mallery; Andrew S. Holpuch; Steven P. Schwendeman

Collaboration


Dive into the Andrew S. Holpuch's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Meng Tong

Ohio State University

View shared research outputs
Top Co-Authors

Avatar

Ping Pei

Ohio State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge