Network


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

Hotspot


Dive into the research topics where Shantanu Balkundi is active.

Publication


Featured researches published by Shantanu Balkundi.


AIDS | 2012

Long-acting nanoformulated antiretroviral therapy elicits potent antiretroviral and neuroprotective responses in HIV-1-infected humanized mice.

Prasanta K. Dash; Howard E. Gendelman; Upal Roy; Shantanu Balkundi; Yazen Alnouti; R.L. Mosley; Harris A. Gelbard; JoEllyn McMillan; Santhi Gorantla; Larisa Y. Poluektova

Objectives:Long-acting nanoformulated antiretroviral therapy (nanoART) with improved pharmacokinetics, biodistribution and limited systemic toxicities will likely improve drug adherence and access to viral reservoirs. Design:Atazanavir and ritonavir crystalline nanoART were formulated in a poloxamer-188 excipient by high-pressure homogenization. These formulations were evaluated for antiretroviral and neuroprotective activities in humanized NOD/scid-IL-2Rgcnull (NSG) mice. Methods:NanoART-treated NSG mice were evaluated for drug biodistribution, pharmacodynamics and toxicity. CD34+ human hematopoietic stem cells were transplanted at birth in replicate NSG mice. The mice were infected with HIV-1ADA at 5 months of age. Eight weeks later, the infected animals were treated with weekly subcutaneous injections of nanoformulated ATV and RTV. Peripheral viral load, CD4+ T-cell counts and lymphoid and brain histopathology and immunohistochemistry tests were performed. Results:NanoART treatments by once-a-week injections reduced viral loads more than 1000-fold and protected CD4+ T-cell populations. This paralleled high ART levels in liver, spleen and blood that were in or around the human minimal effective dose concentration without notable toxicities. Importantly, examination of infected brain subregions showed that nanoART elicited neuroprotective responses with detectable increases in microtubule-associated protein-2, synaptophysin and neurofilament expression when compared to untreated virus-infected animals. Therapeutic interruptions produced profound viral rebounds. Conclusion:Long-acting nanoART has translational potential with sustained and targeted efficacy and with limited systemic toxicities. Such success in drug delivery and distribution could improve drug adherence and reduce viral resistance in infected people.


Nanomedicine: Nanotechnology, Biology and Medicine | 2013

Macrophage folate receptor-targeted antiretroviral therapy facilitates drug entry, retention, antiretroviral activities and biodistribution for reduction of human immunodeficiency virus infections

Pavan Puligujja; JoEllyn McMillan; Lindsey M. Kendrick; Tianyuzi Li; Shantanu Balkundi; Nathan Smith; Ram S. Veerubhotla; Benson Edagwa; Alexander V. Kabanov; Tatiana K. Bronich; Howard E. Gendelman; Xin Ming Liu

UNLABELLED Macrophages serve as vehicles for the carriage and delivery of polymer-coated nanoformulated antiretroviral therapy (nanoART). Although superior to native drug, high drug concentrations are required for viral inhibition. Herein, folate-modified ritonavir-boosted atazanavir (ATV/r)-encased polymers facilitated macrophage receptor targeting for optimizing drug dosing. Folate coating of nanoART ATV/r significantly enhanced cell uptake, retention and antiretroviral activities without altering cell viability. Enhanced retentions of folate-coated nanoART within recycling endosomes provided a stable subcellular drug depot. Importantly, up to a five-fold enhanced plasma and tissue drug levels followed folate-coated formulation injection in mice. Folate polymer encased ATV/r improves nanoART pharmacokinetics bringing the technology one step closer to human use. FROM THE CLINICAL EDITOR This team of authors describes a novel method for macrophage folate receptor-targeted antiretroviral therapy. Atazanvir entry, retention, and antiretroviral activities were superior using the presented method, and so was its biodistribution, enabling a more efficient way to address human immunodeficiency virus infections, with a hoped for clinical application in the near future.


The Journal of Infectious Diseases | 2012

Pharmacodynamic and Antiretroviral Activities of Combination Nanoformulated Antiretrovirals in HIV-1–Infected Human Peripheral Blood Lymphocyte–Reconstituted Mice

Upal Roy; JoEllyn McMillan; Yazen Alnouti; Nagsen Gautum; Nathan Smith; Shantanu Balkundi; Prasanta K. Dash; Santhi Gorantla; Andrea Martinez-Skinner; Jane L. Meza; Georgette D. Kanmogne; Susan Swindells; Samuel M. Cohen; R. Lee Mosley; Larisa Y. Poluektova; Howard E. Gendelman

Lack of adherence, inaccessibility to viral reservoirs, long-term drug toxicities, and treatment failures are limitations of current antiretroviral therapy (ART). These limitations lead to increased viral loads, medicine resistance, immunocompromise, and comorbid conditions. To this end, we developed long-acting nanoformulated ART (nanoART) through modifications of existing atazanavir, ritonavir, and efavirenz suspensions in order to establish cell and tissue drug depots to achieve sustained antiretroviral responses. NanoARTs abilities to affect immune and antiviral responses, before or following human immunodeficiency virus type 1 infection were tested in nonobese severe combined immune-deficient mice reconstituted with human peripheral blood lymphocytes. Weekly subcutaneous injections of drug nanoformulations at doses from 80 mg/kg to 250 mg/kg, 1 day before and/or 1 and 7 days after viral exposure, elicited drug levels that paralleled the human median effective concentration, and with limited toxicities. NanoART treatment attenuated viral replication and preserved CD4(+) Tcell numbers beyond that seen with orally administered native drugs. These investigations bring us one step closer toward using long-acting antiretrovirals in humans.


Antimicrobial Agents and Chemotherapy | 2013

Preclinical Pharmacokinetics and Tissue Distribution of Long-Acting Nanoformulated Antiretroviral Therapy

Nagsen Gautam; Upal Roy; Shantanu Balkundi; Pavan Puligujja; Dongwei Guo; Nathan Smith; Xin Ming Liu; Benjamin G. Lamberty; Brenda Morsey; Howard S. Fox; JoEllyn McMillan; Howard E. Gendelman; Yazen Alnouti

ABSTRACT Long-acting injectable nanoformulated antiretroviral therapy (nanoART) was developed with the explicit goal of improving medicine compliance and for drug targeting of viral tissue reservoirs. Prior nanoART studies completed in humanized virus-infected mice demonstrated sustained antiretroviral responses. However, the pharmacokinetics (PK) and tissue distribution of nanoART were not characterized. To this end, the PK and tissue distribution of nanoformulated atazanavir (ATV) and ritonavir (RTV) injected subcutaneously or intramuscularly in mice and monkeys were evaluated. Fourteen days after injection, ATV and RTV levels were up to 13-, 41-, and 4,500-fold higher than those resulting from native-drug administration in plasma, tissues, and at the site of injection, respectively. At nanoART doses of 10, 50, 100, and 250 mg/kg of body weight, relationships of more- and less-than-proportional increases in plasma and tissue levels with dose increases were demonstrated with ATV and RTV. Multiple-dose regimens showed serum and tissue concentrations up to 270-fold higher than native-drug concentrations throughout 8 weeks of study. Importantly, nanoART was localized in nonlysosomal compartments in tissue macrophages, creating intracellular depot sites. Reflective data were obtained in representative rhesus macaque studies. We conclude that nanoART demonstrates blood and tissue antiretroviral drug levels that are enhanced compared to those of native drugs. The sustained and enhanced PK profile of nanoART is, at least in part, the result of the sustained release of ATV and RTV from tissue macrophases and at the site of injection.


International Journal of Nanomedicine | 2012

Mononuclear phagocyte intercellular crosstalk facilitates transmission of cell-targeted nanoformulated antiretroviral drugs to human brain endothelial cells.

Georgette D. Kanmogne; Sangya Singh; Upal Roy; Xinming Liu; JoEllyn McMillan; Santhi Gorantla; Shantanu Balkundi; Nathan Smith; Yazen Alnouti; Nagsen Gautam; You Zhou; Larisa Y. Poluektova; Alexander V. Kabanov; Tatiana K. Bronich; Howard E. Gendelman

Despite the successes of antiretroviral therapy (ART), HIV-associated neurocognitive disorders remain prevalent in infected people. This is due, in part, to incomplete ART penetration across the blood–brain barrier (BBB) and lymph nodes and to the establishment of viral sanctuaries within the central nervous system. In efforts to improve ART delivery, our laboratories developed a macrophage-carriage system for nanoformulated crystalline ART (nanoART) (atazanavir, ritonavir, indinavir, and efavirenz). We demonstrate that nanoART transfer from mononuclear phagocytes (MP) to human brain microvascular endothelial cells (HBMEC) can be realized through cell-to-cell contacts, which can facilitate drug passage across the BBB. Coculturing of donor MP containing nanoART with recipient HBMEC facilitates intercellular particle transfer. NanoART uptake was observed in up to 52% of HBMEC with limited cytotoxicity. Folate coating of nanoART increased MP to HBMEC particle transfer by up to 77%. To translate the cell assays into relevant animal models of disease, ritonavir and atazanavir nanoformulations were injected into HIV-1-infected NOD/scid-γcnull mice reconstituted with human peripheral blood lymphocytes. Atazanavir and ritonavir levels in brains of mice treated with folate-coated nanoART were three- to four-fold higher than in mice treated with noncoated particles. This was associated with decreased viral load in the spleen and brain, and diminished brain CD11b-associated glial activation. We postulate that monocyte-macrophage transfer of nanoART to brain endothelial cells could facilitate drug entry into the brain.


Biomaterials | 2015

Pharmacodynamics of long-acting folic acid-receptor targeted ritonavir boosted atazanavir nanoformulations

Pavan Puligujja; Shantanu Balkundi; Lindsey M. Kendrick; Hannah M. Baldridge; James Hilaire; Aditya N. Bade; Prasanta K. Dash; Gang Zhang; Larisa Y. Poluektova; Santhi Gorantla; Xin Ming Liu; Tianlei Ying; Yang Feng; Yanping Wang; Dimiter S. Dimitrov; JoEllyn McMillan; Howard E. Gendelman

Long-acting nanoformulated antiretroviral therapy (nanoART) that targets monocyte-macrophages could improve the drugs half-life and protein-binding capacities while facilitating cell and tissue depots. To this end, ART nanoparticles that target the folic acid (FA) receptor and permit cell-based drug depots were examined using pharmacokinetic and pharmacodynamic (PD) tests. FA receptor-targeted poloxamer 407 nanocrystals, containing ritonavir-boosted atazanavir (ATV/r), significantly increased drug bioavailability and PD by five and 100 times, respectively. Drug particles administered to human peripheral blood lymphocyte reconstituted NOD.Cg-Prkdc(scid)Il2rg(tm1Wjl)/SzJ mice and infected with HIV-1ADA led to ATV/r drug concentrations that paralleled FA receptor beta staining in both the macrophage-rich parafollicular areas of spleen and lymph nodes. Drug levels were higher in these tissues than what could be achieved by either native drug or untargeted nanoART particles. The data also mirrored potent reductions in viral loads, tissue viral RNA and numbers of HIV-1p24+ cells in infected and treated animals. We conclude that FA-P407 coating of ART nanoparticles readily facilitates drug carriage and antiretroviral responses.


International Journal of Nanomedicine | 2011

Comparative manufacture and cell-based delivery of antiretroviral nanoformulations.

Shantanu Balkundi; Ari S. Nowacek; Ram S. Veerubhotla; Han Chen; Andrea Martinez-Skinner; Upal Roy; R. Lee Mosley; Georgette D. Kanmogne; Xinming Liu; Alexander V. Kabanov; Tatiana K. Bronich; JoEllyn McMillan; Howard E. Gendelman

Nanoformulations of crystalline indinavir, ritonavir, atazanavir, and efavirenz were manufactured by wet milling, homogenization or sonication with a variety of excipients. The chemical, biological, immune, virological, and toxicological properties of these formulations were compared using an established monocyte-derived macrophage scoring indicator system. Measurements of drug uptake, retention, release, and antiretroviral activity demonstrated differences amongst preparation methods. Interestingly, for drug cell targeting and antiretroviral responses the most significant difference among the particles was the drug itself. We posit that the choice of drug and formulation composition may ultimately affect clinical utility.


Nanotoxicology | 2011

Pharmacotoxicology of monocyte-macrophage nanoformulated antiretroviral drug uptake and carriage

Rafael Bressani; Ari S. Nowacek; Sangya Singh; Shantanu Balkundi; Barrett Rabinow; JoEllyn McMillan; Howard E. Gendelman; Georgette D. Kanmogne

Abstract Limitations inherent to antiretroviral therapy (ART) in its pharmacokinetic properties remain despite over 15 years of broad use. Our laboratory has pioneered a means to improve ART delivery through monocyte-macrophage carriage of nanoformulated drug-encapsulated particles (nanoART). To this end, our prior works sought to optimize nanoART size, charge, and physical properties for cell uptake and antiretroviral activities. To test the functional consequences of indinavir, ritonavir, and efavirenz formulations we investigated relationships between human monocyte and macrophage cytotoxicities and nanoART dose, size, surfactant, and preparation. Wet-milled particles were more cytotoxic to monocytes-macrophages than those prepared by homogenization; with concurrent induction of tumor necrosis factor-alpha. Interestingly, pure suspensions of indinavir and ritonavir at 0.5 mM, and efavirenz at 0.1 mM and 0.5 mM also proved cytotoxic. Individual surfactants and formulated fluconazole neither affected cell function or viability. Although nanoART did not alter brain tight junction proteins ZO-2 and occludin, 0. 5mM ritonavir formulations did alter brain transendothelial electric resistance. These results underscore the potential importance of evaluating the physicochemical and functional properties of nanoART before human evaluations.


Antimicrobial Agents and Chemotherapy | 2014

Pharmacokinetics, Biodistribution, and Toxicity of Folic Acid-Coated Antiretroviral Nanoformulations

Nagsen Gautam; Pavan Puligujja; Shantanu Balkundi; Rhishikesh Thakare; Xin Ming Liu; Howard S. Fox; JoEllyn McMillan; Howard E. Gendelman; Yazen Alnouti

ABSTRACT The drug delivery platform for folic acid (FA)-coated nanoformulated ritonavir (RTV)-boosted atazanavir (FA-nanoATV/r) using poloxamer 407 was developed to enhance cell and tissue targeting for a range of antiretroviral drugs. Such formulations would serve to extend the drug half-life while improving the pharmacokinetic profile and biodistribution to reservoirs of human immunodeficiency virus (HIV) infection. To this end, we now report enhanced pharmacokinetics and drug biodistribution with limited local and systemic toxicities of this novel nanoformulation. The use of FA as a targeting ligand for nanoATV/r resulted in plasma and tissue drug concentrations up to 200-fold higher compared to equimolar doses of native drug. In addition, ATV and RTV concentrations in plasma from mice on a folate-deficient diet were up to 23-fold higher for mice administered FA-nanoATV/r than for mice on a normal diet. Compared to earlier nanoATV/r formulations, FA-nanoATV/r resulted in enhanced and sustained plasma and tissue ATV concentrations. In a drug interaction study, ATV plasma and tissue concentrations were up to 5-fold higher in mice treated with FA-nanoATV/r than in mice treated with FA-nanoATV alone. As observed in mice, enhanced and sustained plasma concentrations of ATV were observed in monkeys. NanoATV/r was associated with transient local inflammation at the site of injection. There were no systemic adverse reactions associated with up to 10 weeks of chronic exposure of mice or monkeys to FA-nanoATV/r.


Journal of Visualized Experiments | 2010

Methods Development for Blood Borne Macrophage Carriage of Nanoformulated Antiretroviral Drugs

Shantanu Balkundi; Ari S. Nowacek; Upal Roy; Andrea Martinez-Skinner; JoEllyn McMillan; Howard E. Gendelman

Nanoformulated drugs can improve pharmacodynamics and bioavailability while serving also to reduce drug toxicities for antiretroviral (ART) medicines. To this end, our laboratory has applied the principles of nanomedicine to simplify ART regimens and as such reduce toxicities while improving compliance and drug pharmacokinetics. Simple and reliable methods for manufacturing nanoformulated ART (nanoART) are shown. Particles of pure drug are encapsulated by a thin layer of surfactant lipid coating and produced by fractionating larger drug crystals into smaller ones by either wet milling or high-pressure homogenization. In an alternative method free drug is suspended in a droplet of a polymer. Herein, drug is dissolved within a polymer then agitated by ultrasonication until individual nanosized droplets are formed. Dynamic light scattering and microscopic examination characterize the physical properties of the particles (particle size, charge and shape). Their biologic properties (cell uptake and retention, cytotoxicity and antiretroviral efficacy) are determined with human monocyte-derived macrophages (MDM). MDM are derived from human peripheral blood monocytes isolated from leukopacks using centrifugal elutriation for purification. Such blood-borne macrophages may be used as cellular transporters for nanoART distribution to human immunodeficiency virus (HIV) infected organs. We posit that the repackaging of clinically available antiretroviral medications into nanoparticles for HIV-1 treatments may improve compliance and positively affect disease outcomes.

Collaboration


Dive into the Shantanu Balkundi's collaboration.

Top Co-Authors

Avatar

Howard E. Gendelman

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

JoEllyn McMillan

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Upal Roy

Florida International University

View shared research outputs
Top Co-Authors

Avatar

Yazen Alnouti

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Georgette D. Kanmogne

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Pavan Puligujja

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alexander V. Kabanov

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Andrea Martinez-Skinner

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ari S. Nowacek

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Larisa Y. Poluektova

University of Nebraska Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge