Network


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

Hotspot


Dive into the research topics where Prashanth K.B. Nagesh is active.

Publication


Featured researches published by Prashanth K.B. Nagesh.


Aaps Journal | 2015

Therapeutic Applications of Curcumin Nanoformulations

Murali M. Yallapu; Prashanth K.B. Nagesh; Meena Jaggi; Subhash C. Chauhan

Curcumin (diferuloylmethane) is a bioactive and major phenolic component of turmeric derived from the rhizomes of curcuma longa linn. For centuries, curcumin has exhibited excellent therapeutic benefits in various diseases. Owing to its anti-oxidant and anti-inflammatory properties, curcumin plays a significant beneficial and pleiotropic regulatory role in various pathological conditions including cancer, cardiovascular disease, Alzheimer’s disease, inflammatory disorders, neurological disorders, and so on. Despite such phenomenal advances in medicinal applications, the clinical implication of native curcumin is hindered due to low solubility, physico-chemical instability, poor bioavailability, rapid metabolism, and poor pharmacokinetics. However, these issues can be overcome by utilizing an efficient delivery system. Active scientific research was initiated in 2005 to improve curcumin’s pharmacokinetics, systemic bioavailability, and biological activity by encapsulating or by loading curcumin into nanoform(s) (nanoformulations). A significant number of nanoformulations exist that can be translated toward medicinal use upon successful completion of pre-clinical and human clinical trials. Considering this perspective, current review provides an overview of an efficient curcumin nanoformulation for a targeted therapeutic option for various human diseases. In this review article, we discuss the clinical evidence, current status, and future opportunities of curcumin nanoformulation(s) in the field of medicine. In addition, this review presents a concise summary of the actions required to develop curcumin nanoformulations as pharmaceutical or nutraceutical candidates.


Colloids and Surfaces B: Biointerfaces | 2016

PSMA targeted docetaxel-loaded superparamagnetic iron oxide nanoparticles for prostate cancer.

Prashanth K.B. Nagesh; Nia R. Johnson; Vijaya K.N. Boya; Pallabita Chowdhury; Shadi F. Othman; Vahid Khalilzad-Sharghi; Bilal B. Hafeez; Aditya Ganju; Sheema Khan; Stephen W. Behrman; Nadeem Zafar; Subhash C. Chauhan; Meena Jaggi; Murali M. Yallapu

Docetaxel (Dtxl) is currently the most common therapeutic option for prostate cancer (PC). However, adverse side effects and problems associated with chemo-resistance limit its therapeutic outcome in clinical settings. A targeted nanoparticle system to improve its delivery to and activity at the tumor site could be an attractive strategy for PC therapy. Therefore, the objective of this study was to develop and determine the anti-cancer efficacy of a novel docetaxel loaded, prostate specific membrane antigen (PSMA) targeted superparamagnetic iron oxide nanoparticle (SPION) (J591-SPION-Dtxl) formulation for PC therapy. Our results showed the SPION-Dtxl formulation exhibits an optimal particle size and zeta potential, which can efficiently be internalized in PC cells. SPION-Dtxl exhibited potent anti-cancer efficacy via induction of the expression of apoptosis associated proteins, downregulation of anti-apoptotic proteins, and inhibition of chemo-resistance associated protein in PC cell lines. J591-SPION-Dtxl exhibited a profound uptake in C4-2 (PSMA(+)) cells compared to PC-3 (PSMA(-)) cells. A similar targeting potential was observed in ex-vivo studies in C4-2 tumors but not in PC-3 tumors, suggesting its tumor specific targeting. Overall, this study suggests that a PSMA antibody functionalized SPION-Dtxl formulation can be highly useful for targeted PC therapy.


Journal of Colloid and Interface Science | 2017

Probing mucin interaction behavior of magnetic nanoparticles

Vijayakumar N. Boya; Renn Lovett; Saini Setua; Vaibhav Gandhi; Prashanth K.B. Nagesh; Sheema Khan; Meena Jaggi; Murali M. Yallapu; Subhash C. Chauhan

In this study, we developed iron oxide based magnetic nanoparticles (MNPs) by precipitation of iron salts in the presence of ammonia and created four different formulations: without functionality (plain MNPs, no coating), with β-cyclodextrin (MNPs+β-CD) or pluronic 127 polymer (MNPs+F-127), and both β-cyclodextrin and pluronic 127 polymer (MNPs+β-CD-F-127) functionality for its efficient use in mucosal delivery. We studied the interaction and/or binding behavior of these MNPs formulations with porcine stomach mucin using steady-state fluorescence spectroscopy, and then quantified the bound mucin from absorption studies. Toxicity of these MNPs against cervical cancer cells and red blood cells was evaluated. Ex-vivo studies were performed using freshly collected gastrointestinal, ovarian, pancreas and colon organ tissues of pig to evaluate binding and uptake phenomenon of MNPs. Transport studies of these MNPs in mucin was evaluated using Boydens chamber assay. All these studies together suggest that the MNPs+β-CD-F-127 formulation was strongly interacted with mucin and interestingly transported through mucin compared to other MNPs formulations. Hence, MNPs+β-CD-F-127 formulation could be a good candidate for the mucoadhesive biopharmaceuticals and drug delivery system.


Acta Pharmaceutica Sinica B | 2017

Development of polyvinylpyrrolidone/paclitaxel self-assemblies for breast cancer

Pallabita Chowdhury; Prashanth K.B. Nagesh; Sheema Khan; Bilal B. Hafeez; Subhash C. Chauhan; Meena Jaggi; Murali M. Yallapu

The goal of this investigation was to develop and demonstrate a polymer/paclitaxel self-assembly (PTX-SA) formulation. Polymer/PTX-SAs were screened based on smaller size of formulation using dynamic light scattering analysis. Additionally, fluorescence microscopy and flow cytometry studies exhibited that polyvinylpyrrolidone (PVP)-based PTX-SAs (PVP/PTX-SAs) had superior cellular internalization capability in MCF7 and MDA-MB-231 breast cancer cells. The optimized PVP/PTX-SAs exhibited less toxicity to human red blood cells indicating a suitable formulation for reducing systemic toxicity. The formation of PVP and PTX self-assemblies was confirmed using fluorescence quenching and transmission electron microscopy which indicated that the PVP/PTX-SAs were spherical in shape with an average size range of 53.81 nm as detected by transmission electron microscopy (TEM). FTIR spectral analysis demonstrates incorporation of polymer and paclitaxel functional groups in PVP/PTX-SAs. Both proliferation (MTS) and clonogenic (colony formation) assays were used to validate superior anticancer activity of PVP/PTX-SAs in breast cancer cells over paclitaxel. Such superior anticancer activity was also demonstrated by downregulation of the expression of pro-survival protein (Bcl-xL), upregulation of apoptosis-associated proteins (Bid, Bax, cleaved caspase 7, and cleaved PARP) and β-tubulin stabilization. These results support the hypothesis that PVP/PTX-SAs improved paclitaxel delivery to cancer cells.


Archive | 2017

Pluronic Nanotechnology for Overcoming Drug Resistance

Pallabita Chowdhury; Prashanth K.B. Nagesh; Santosh Kumar; Meena Jaggi; Subhash C. Chauhan; Murali M. Yallapu

Chemotherapy is one of the most conventionally used therapeutic interventions for treating various diseases. Chances of acquiring multidrug resistance in response to chemotherapeutic agents are exceedingly common among patients. Drug resistance arises mainly due to overexpression of efflux transporters such as P-glycoprotein and multidrug resistance-associated protein of the ATP-binding cassette superfamily of proteins, which significantly limits intracellular drug accumulation and drug activity. Although many approaches exist to overcome drug resistance, their uses are significantly limited in clinical practice. In this chapter, we demonstrate the superior functions of Pluronic-based technologies to overcome drug resistance. The present chapter highlights various aspects of Pluronic polymers, Pluronic conjugates, Pluronic nanotechnology, as well as their therapeutic implications for effective treatment strategies. We include the role of Pluronic polymers as a pharmaceutic excipient and drug delivery vehicle in this review. In addition, we highlight examples of Pluronic nanosystems that are currently in preclinical development, clinical trials, and clinically translatable formulations. Furthermore, a number of innovative Pluronic nano-designs of advanced therapeutics for future medicinal applications are presented. Collectively, the use of Pluronic-based nanoformulations discussed in this chapter suggests sensitization and prevention of drug resistance. Such an approach not only minimizes the dose required for treatment, but also minimizes the number of treatment cycles, which is useful in a clinical scenario.


Cancer Research | 2016

Abstract 1312: PSMA antibody functionalized docetaxel-loaded magnetic nanoparticles for prostate cancer therapy

Prashanth K.B. Nagesh; Nia R. Johnson; Vijaya K.N. Boya; Pallabita Chowdhury; Aditya Ganju; Bilal B. Hafeez; Sheema Khan; Meena Jaggi; Subhash C. Chauhan; Murali M. Yallapu

Objectives: Prostate cancer (PrCa) is the second most leading cause of cancer-related death in men in the United States. Chemotherapy (Docetaxel, Dox) is currently the most common first-line therapeutic option. However, adverse side effects and chemo-resistance of docetaxel limit its clinical use. Improving docetaxel targeted delivery and its activity at the tumor site using a targeted nanoparticle system could be an attractive strategy for PrCa therapy. Prostate Specific Membrane Antigen (PSMA) is highly overexpressed in PrCa cells, thus is a highly attractive molecular target for PrCa therapy. In this study, we developed and determined anti-cancer efficacy of a novel docetaxel loaded, PSMA targeted magnetic nanoparticle (PSMA-MNP-Dox) formulation for PrCa therapy. Methods: Docetaxel loaded magnetic nanoparticle (MNP-Dox) formulation is composed of an iron oxide core coated with cyclodextrin (for drug loading) and F127 polymer (for particle stability and chemosensitization). Therapeutic efficacy of this unique nanoparticle formulation was evaluated using clinically relevant cell line models (C4-2, PC-3, and DU-145) through cell proliferation and colony formation assays. Molecular effects of this formulation on apoptosis, anti-apotosis, and drug resistance associated proteins were evaluated using immunoblotting assays. Contrast imaging property of MNP-Dox formulation was examined using Phantom Gel MR imaging model. For active targeting, PSMA antibody conjugation to this formulation was achieved through N-hydroxysuccinimide group containing PEG polymer. Active targeting potential of this formulation was evaluated in PSMA+ (C4-2) and PSMA- (PC-3) cell lines, C4-2 generated tumor xenografts. Results: MNP-Dox formulation showed optimal particle size and zeta potential which can efficiently internalized in PrCa cells. Our formulation showed anti-cancer efficacy in prostate cancer cell lines. Additionally, it induces the expression of apoptosis associated proteins, Bax and Bad, cleaved PARP, and caspase 3, and downregulated the expression of anti-apoptotic proteins, Bcl-2 and Bcl-xL. Moreover, it also inhibited the expression of chemoresistance associated proteins (PSMA and MDR1). Our PSMA antibody targeted MNPs-Dox formulation exhibited a profound uptake pattern in PSMA+ cells (C4-2) compared to PSMA null (PC-3)- cells, suggesting its targeting potential. A similar targeting potential was also observed in ex-vivo studies while using C4-2 tumor xenografts, however, no intense targeting was observed in normal tissues due to lack of PSMA expression. Conclusion: PSMA antibody functionalized MNP-Dox formulation can efficiently target PSMA + PrCa cells and deliver docetaxel into prostate tumors. This targeted drug delivery system could reduce the dose of docetaxel required to kill cancer cells, thus minimizing long-term docetaxel associated systemic toxicity and drug-resistance. Citation Format: Prashanth Kumar Bhusetty Nagesh, Nia Johnson, Vijaya K.N. Boya, Pallabita Chowdhury, Aditya Ganju, Bilal Hafeez, Sheema Khan, Meena Jaggi, Subhash C. Chauhan, Murali M. Yallapu. PSMA antibody functionalized docetaxel-loaded magnetic nanoparticles for prostate cancer therapy. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1312.


Cancers | 2018

Tannic acid induces endoplasmic reticulum stress-mediated apoptosis in prostate cancer

Prashanth K.B. Nagesh; Elham Hatami; Pallabita Chowdhury; Vivek K. Kashyap; Sheema Khan; Bilal B. Hafeez; Subhash C. Chauhan; Meena Jaggi; Murali M. Yallapu

Endoplasmic reticulum (ER) stress is an intriguing target with significant clinical importance in chemotherapy. Interference with ER functions can lead to the accumulation of unfolded proteins, as detected by transmembrane sensors that instigate the unfolded protein response (UPR). Therefore, controlling induced UPR via ER stress with natural compounds could be a novel therapeutic strategy for the management of prostate cancer. Tannic acid (a naturally occurring polyphenol) was used to examine the ER stress mediated UPR pathway in prostate cancer cells. Tannic acid treatment inhibited the growth, clonogenic, invasive, and migratory potential of prostate cancer cells. Tannic acid demonstrated activation of ER stress response (Protein kinase R-like endoplasmic reticulum kinase (PERK) and inositol requiring enzyme 1 (IRE1)) and altered its regulatory proteins (ATF4, Bip, and PDI) expression. Tannic acid treatment affirmed upregulation of apoptosis-associated markers (Bak, Bim, cleaved caspase 3, and cleaved PARP), while downregulation of pro-survival proteins (Bcl-2 and Bcl-xL). Tannic acid exhibited elevated G1 population, due to increase in p18INK4C and p21WAF1/CIP1 expression, while cyclin D1 expression was inhibited. Reduction of MMP2 and MMP9, and reinstated E-cadherin signifies the anti-metastatic potential of this compound. Altogether, these results demonstrate that tannic acid can promote apoptosis via the ER stress mediated UPR pathway, indicating a potential candidate for cancer treatment.


Journal of Colloid and Interface Science | 2019

Tannic acid-inspired paclitaxel nanoparticles for enhanced anticancer effects in breast cancer cells

Pallabita Chowdhury; Prashanth K.B. Nagesh; Elham Hatami; Santosh Wagh; Nirnoy Dan; Manish K. Tripathi; Sheema Khan; Bilal Bin Hafeez; Bernd Meibohm; Subhash C. Chauhan; Meena Jaggi; Murali M. Yallapu

Paclitaxel (PTX) is a gold standard chemotherapeutic agent for breast, ovarian, pancreatic and non-small cell lung carcinoma. However, in clinical use PTX can have adverse side effects or inadequate pharmacodynamic parameters, limiting its use. Nanotechnology is often employed to reduce the therapeutic dosage required for effective therapy, while also minimizing the systemic side effects of chemotherapy drugs. However, there is no nanoformulation of paclitaxel with chemosensitization motifs built in. With this objective, we screened eleven pharmaceutical excipients to develop an alternative paclitaxel nanoformulation using a self-assembly method. Based on the screening results, we observed tannic acid possesses unique properties to produce a paclitaxel nanoparticle formulation, i.e., tannic acid-paclitaxel nanoparticles. This stable TAP nanoformulation, referred to as TAP nanoparticles (TAP NPs), showed a spherical shape of ~ 102 nm and negative zeta potential of ~ -8.85. The presence of PTX in TAP NPs was confirmed by Fourier Transform Infrared (FTIR) spectra, thermogravimetric analyzer (TGA), and X-ray diffraction (XRD). Encapsulation efficiency of PTX in TAP NPs was determined to be ≥96%. Intracellular drug uptake of plain drug PTX on breast cancer cells (MDA-MB-231) shows more or less constant drug levels in 2 to 6 h, suggesting drug efflux by the P-gp transporters, over TAP NPs, in which PTX uptake was more than 95.52 ± 11.01% in 6 h, as analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Various biological assays such as proliferation, clonogenic formation, invasion, and migration confirm superior anticancer effects of TAP NPs over plain PTX at all tested concentrations. P-gp expression, beta-tubulin stabilization, Western blot, and microarray analysis further confirm the improved therapeutic potential of TAP NPs. These results suggest that the TAP nanoformulation provides an important reference for developing a therapeutic nanoformulation affording pronounced, enhanced effects in breast cancer therapy.


Pharmaceutics | 2018

Tannic Acid-Lung Fluid Assemblies Promote Interaction and Delivery of Drugs to Lung Cancer Cells

Elham Hatami; Prashanth K.B. Nagesh; Pallabita Chowdhury; Subhash C. Chauhan; Meena Jaggi; Amali Samarasinghe; Murali M. Yallapu

Lung cancer (LC) is one of the leading causes of death in both men and women in the United States. Tannic acid (TA), a water-soluble polyphenol, exhibits a wide range of biological activities. TA has received much attention as a promising compound in the biomaterial and drug delivery fields. Lung fluid (LF) is a major barrier for distribution of drugs to the lungs. Therefore, the purpose of this study was to examine TA interaction with LF for effective delivery of anti-cancer drug molecules via pulmonary delivery. The extent of adsorption of LF proteins by TA was revealed by fluorescence quenching in fluorescence spectroscopy. The presence of LF in TA-LF complexes was noticed by the presence of protein peaks at 1653 cm−1. Both protein dot and SDS-PAGE analysis confirmed LF protein complexation at all TA concentrations employed. A stable particle TA-LF complex formation was observed through transmission electron microscopy (TEM) analysis. The complexation pattern measured by dynamic light scattering (DLS) indicated that it varies depending on the pH of the solutions. The degree of LF presence in TA-LF complexes signifies its interactive behavior in LC cell lines. Such superior interaction offered an enhanced anti-cancer activity of drugs encapsulated in TA-LF complex nanoformulations. Our results indicate that TA binds to LF and forms self-assemblies, which profoundly enhance interaction with LC cells. This study demonstrated that TA is a novel carrier for pharmaceutical drugs such as gemcitabine, carboplatin, and irinotecan.


Cancers | 2018

miRNA-205 Nanoformulation Sensitizes Prostate Cancer Cells to Chemotherapy

Prashanth K.B. Nagesh; Pallabita Chowdhury; Elham Hatami; Vijaya K.N. Boya; Vivek K. Kashyap; Sheema Khan; Bilal B. Hafeez; Subhash C. Chauhan; Meena Jaggi; Murali M. Yallapu

The therapeutic application of microRNA(s) in the field of cancer has generated significant attention in research. Previous studies have shown that miR-205 negatively regulates prostate cancer cell proliferation, metastasis, and drug resistance. However, the delivery of miR-205 is an unmet clinical need. Thus, the development of a viable nanoparticle platform to deliver miR-205 is highly sought. A novel magnetic nanoparticle (MNP)-based nanoplatform composed of an iron oxide core with poly(ethyleneimine)-poly(ethylene glycol) layer(s) was developed. An optimized nanoplatform composition was confirmed by examining the binding profiles of MNPs with miR-205 using agarose gel and fluorescence methods. The novel formulation was applied to prostate cancer cells for evaluating cellular uptake, miR-205 delivery, and anticancer, antimetastasis, and chemosensitization potentials against docetaxel treatment. The improved uptake and efficacy of formulations were studied with confocal imaging, flow cytometry, proliferation, clonogenicity, Western blot, q-RT-PCR, and chemosensitization assays. Our findings demonstrated that the miR-205 nanoplatform induces significant apoptosis and enhancing chemotherapeutic effects in prostate cancer cells. Overall, these study results provide a strong proof-of-concept for a novel nonviral-based nanoparticle protocol for effective microRNA delivery to prostate cancer cells.

Collaboration


Dive into the Prashanth K.B. Nagesh's collaboration.

Top Co-Authors

Avatar

Meena Jaggi

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Murali M. Yallapu

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Subhash C. Chauhan

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Pallabita Chowdhury

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Sheema Khan

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Elham Hatami

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Bilal Bin Hafeez

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Bilal B. Hafeez

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Vivek K. Kashyap

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Aditya Ganju

University of Tennessee Health Science Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge