Department of Pharmaceutics

 

Full publication list/research output can be viewed at 

https://manipal.pure.elsevier.com/en/organisations/department-of-pharmaceutics-manipal-college-of-pharmaceutical-sci

 

    2021

 

1.      Pandey, A., Nikam, A. N., Fernandes, G., Kulkarni, S., Padya, B. S., Prassl, R., Das, S.,  Joseph, A., Deshmukh, P. K., Patil, P. O. & Mutalik, S

Black phosphorus as multifaceted advanced material nanoplatforms for potential biomedical applications

Nanomaterials 2021, 11(1), 13.

Black phosphorus is one of the emerging members of two-dimen-sional (2D) materials which has recently entered the biomedical field. Its anisotropic properties and infrared bandgap have enabled researchers to discover its applicability in several fields including optoelectronics, 3D printing, bioim-aging, and others. Characterization techniques such as Raman spectroscopy have revealed the structural information of Black phosphorus (BP) along with its fundamental properties, such as the behavior of its photons and electrons. The present review provides an overview of synthetic approaches and properties of BP, in addition to a detailed discussion about various types of surface modifications available for overcoming the stability-related drawbacks and for imparting targeting ability to synthesized nanoplatforms. The review further gives an overview of multiple characterization techniques such as spectro-scopic, thermal, optical, and electron microscopic techniques for providing an insight into its fundamental properties. These characterization techniques are not only important for the analysis of the synthesized BP but also play a vital role in assessing the doping as well as the structural integrity of BP-based nano-composites. The potential role of BP and BP-based nanocomposites for biomed-ical applications specifically, in the fields of drug delivery, 3D printing, and wound dressing, have been discussed in detail to provide an insight into the multifunctional role of BP-based nanoplatforms for the management of various diseases, including cancer therapy. The review further sheds light on the role of BP-based 2D platforms such as BP nanosheets along with BP-based 0D plat-forms—i.e., BP quantum dots in the field of therapy and bioimaging of cancer using techniques such as photoacoustic imaging and fluorescence imaging. Alt-hough the review inculcates the multimodal therapeutic as well as imaging role of BP, there is still research going on in this field which will help in the development of BP-based theranostic platforms not only for cancer therapy, but various other diseases.

2.      Narayan, R., Gadag, S., Cheruku, S. P., Raichur, A. M., Day, C. M., Garg, S., Manandhar, S., Pai, K. S. R., Suresh, A., Mehta, C. H., Nayak, Y., Kumar, N. & Nayak, U. Y.

Chitosan-glucuronic acid conjugate coated mesoporous silica nanoparticles: A smart pH-responsive and receptor-targeted system for colorectal cancer therapy

Carbohydrate Polymers, 261, 2021, 117893

Glycosylated pH-sensitive mesoporous silica nanoparticles (MSNs) of capecitabine (CAP) were developed for targeting colorectal cancer. The MSNs possessed an average pore diameter of 8.12 ± 0.43 nm, pore volume of 0.73 ± 0.21 cm3/g, and particle size of 245.24 ± 5.75 nm. A high loading of 180.51 ± 5.23 mg/g attributed to the larger pore volume was observed. The surface of the drug-loaded MSNs were capped with chitosan-glucuronic acid (CHS-GCA) conjugate to combine two strategies viz. pH-sensitive, and lectin receptor mediated uptake. In vitro studies demonstrated a pH-sensitive and controlled release of CAP which was further enhanced in the presence of rat caecal content. Higher uptake of the (CAP-MSN)CHS-GCA was observed in HCT 116 cell lines. The glycosylated nanoparticles revealed reduction in the tumors, aberrant crypt foci, dysplasia and inflammation, and alleviation in the toxic features. This illustrated that the nanoparticles showed promising antitumor efficacy with reduced toxicity and may be used as a effective carrier against cancer.

3.      Bhaskaran, N. A., Kumar, L., Reddy, M. S. & Pai, G. K.

An analytical "quality by design" approach in RP-HPLC method development and validation for reliable and rapid estimation of irinotecan in an injectable formulation

Acta Pharm. 2021, 71(1):57-79.

The objective of the present study was to develop a robust, simple, economical and sensitive HPLC-UV method using the "quality-by-design" approach for the estimation of irinotecan (IRI) in marketed formulations. RP-HPLC method was developed by applying Box-Behnken design with Hyper-Clone (Phenomenex®) C18 column (250 × 4.6 mm id, particle size 5 µm, ODS 130 Å) as a stationary phase. Acetonitrile and 20 mmol L-1 potassium phosphate buffer (pH 2.5) containing 0.1 % triethylamine in a ratio of 45:55 % (V/V) was used as a mobile phase. The sample was injected in a volume of 20 µL into the HPLC system. UV detector at 254 nm was used to estimate and quantify IRI. Isocratic elution was opted while the flow rate was maintained at 0.75 mL min-1. The retention time of IRI was found to be 4.09 min. The responses were found to be linear for concentration range of 0.5 to 18.0 µg mL-1 and the coefficient of determination value was found to be 0.9993. Percent relative standard deviation for intra- and inter-day precisions was found in the range of 0.1 to 0.4 %. LOD and LOQ values were found to be 4.87 and 14.75 ng mL-1, resp. Robustness studies confirmed that the developed method is robust with RSD of a maximum 0.1 %. The method is simple, precise, sensitive, robust and economical making it applicable to the estimation of IRI in an injectable formulation.

 

2020

1.      Chegireddy, M., Hanegave, G. K., Lakshman, D., Urazov, A., Sree, K. N., Lewis, S. A. & Dengale, S. J.

The Significance of Utilizing In Vitro Transfer Model and Media Selection to Study the Dissolution Performance of Weak Ionizable Bases: Investigation Using Saquinavir as a Model Drug

AAPS PharmSciTech. 2020 Jan 3;21(2):47

This study investigated the dissolution behavior of BCS class II ionizable weak base Saquinavir and its mesylate salt in the multi-compartment transfer setup employing different composition of dissolution media. The dissolution behavior of Saquinavir was studied by using a two-compartment transfer model representing the transfer of drug from the stomach (donor compartment) to the upper intestine (acceptor compartment). Various buffers like phosphate, bicarbonate, FaSSIF, and FeSSIF were employed. The dissolution was also studied in the concomitant presence of the additional solute, i.e., Quercetin. Further, the dissolution profiles of Saquinavir and its mesylate salt were simulated by GastroPlusTM, and the simulated dissolution profiles were compared against the experimental ones. The formation of in situ HCl salt and water-soluble amorphous phosphate aggregates was confirmed in the donor and acceptor compartments of the transfer setup, respectively. As the consequence of the lower solubility product of HCl salt of Saquinavir, the solubility advantage of mesylate salt was vanished leading to the lower than the predicted dissolution in the acceptor compartment. However, the formation of water-soluble aggregates in the presence of the phosphate salts was observed leading to the higher than the predicted dissolution of the free base in the transfer setup. Interestingly, the formation of such water-soluble aggregates was found to be hindered in the concomitant presence of an ionic solute resulting in the lower dissolution rates. The in situ generation of salts and aggregates in the transfer model lead to the inconsistent prediction of dissolution profiles by GastroPlusTM.

 

2.      Manikkath, J., Parekh, H. S. & Mutalik, S.

Surface-engineered nanoliposomes with lipidated and non-lipidated peptide-dendrimeric scaffold for efficient transdermal delivery of a therapeutic agent: Development, characterization, toxicological and preclinical performance analyses.

Eur J Pharm Biopharm, 156 (2020), 97-113.

The current study aimed to develop novel peptide dendrimer (PD)-conjugated nanoliposomal formulations of asenapine maleate (ASP) for improvement in the transdermal delivery and pharmacokinetic profile of the drug. Novel arginine-terminated PDs (+/-lipidation) were prepared by solid phase peptide synthesis, followed by conjugation onto ASP nanoliposomes. The nanoliposomes were characterized for particle size (and polydispersity index), zeta potential (ZP), drug entrapment efficiency, shape and morphology, differential scanning calorimetry and FT-IR spectroscopy. Ex vivo skin permeation and retention studies demonstrated considerably higher percutaneous permeation of ASP from the developed nanoliposomes (Q24 = 794.31 ± 54.89 µg/cm2, Jss = 105.40 ± 4.8 µg/cm2/h, ER = 36.85 ± 2.89 for liposomes with lipidated peptide dendrimer (Lipo-PD2)) in comparison with passive diffusion studies (Q24 = 63.09 ± 3.56 µg/cm2, Jss = 3.01 ± 0.23 µg/cm2/h). Confocal Laser Scanning Microscopy (CLSM) confirmed the higher percutaneous penetration of Lipo-PD2 in comparison with liposomes without the dendrimer. In vitro cytotoxicity determined on HaCaT cell line demonstrated CTC50 of >1000 µg/mL for both the synthesized PDs and Lipo-PD2. Pharmacokinetic studies in male Sprague Dawley rats revealed considerably and significantly higher t1/2 = 82.32 ± 14.48 h and AUC0-t = 4403.34 ± 367.10 h.ng/mL, from the developed formulation, compared to orally administered ASP (t1/2 = 21.64 ± 2.53 h and AUC0-t = 2303.55 ± 444.5 h.ng/mL), demonstrating higher bioavailability and longer retention in vivo. Additionally, in vivo skin retention, brain uptake studies and pharmacodynamics of the developed formulations were investigated. Stability studies indicated that the formulations were stable up to relatively stable with respect to size, ZP and drug content for 4 months at the tested conditions. This study demonstrates that the developed PD-conjugated nanoliposomal formulations can effectively serve as a transdermal delivery strategy for ASP.

 

2019

1.  Ghate, Vivek M and Raja, Selvaraj and Lewis, Shaila Angela (2019) 

Development of mart for the rapid production of nanostructured lipid carriers loaded with all-trans retinoic acid for dermal delivery.

AAPS PharmSciTech, 20. pp. 162-178. ISSN 1530-9932

All-trans retinoic acid (ATRA) has been regarded as a wonder drug for many dermatological complications; however, its application is limited due to the extreme irritation, and toxicity seen once it has sufficiently concentrated into the bloodstream from the skin. Thus, the present study was aimed to increase the entrapment of ATRA and minimize its transdermal permeation. ATRA incorporated within nanostructured lipid carriers (NLCs) were produced by a green and facile thin lipid-film based microwave-assisted rapid technique (MART). The optimization was carried out using the response surface methodology (RSM)- driven artificial neural network (ANN) coupled with genetic algorithm (GA). The liquid lipid and surfactants were seen to play a very crucial role culminating in the particle size (< 70 nm), zeta potential (< - 32 mV), and entrapment of ATRA (> 98%). ANN-GA-optimized NLCs required a minimal quantity of the surfactants, formed within 2 min and were stable for 1 year at different storage conditions. The optimized NLC-loaded creams showed a skin retention (ex vivo) to an extent of 87.42% with no detectable drug in the receptor fluid (24 h) in comparison to the marketed cream which released 47.32% (12 h) of ATRA. The results were in good correlation with the in vivo skin deposition studies. The NLCs were biocompatible and non-skin irritant based on the primary irritation index. In conclusion, the NLCs were seen to have a very high potential in overcoming the drawbacks of ATRA for dermal delivery and could be produced conveniently by the MART.

2. Managuli, Renuka S and Raut, Sushil Y and Ajjappla, Shreya B and Mutalik, Srinivas (2019) 

Asenapine maleate-loaded nanostructured lipid carriers: optimization and in vitro, ex vivo and in vivo evaluations.

Nanomedicine, 14 (7). pp. 889-910. ISSN 1743-5889

To prepare nanostructured lipid carriers (NLCs) loaded with asenapine maleate (ASPM) to increase its oral bioavailability by intestinal lymphatic uptake. Materials & methods: ASPM-NLCs were prepared by ultrasound dispersion technique, by adopting Design of Experiment approach, and characterized. Results:The optimized formulation exhibited good physicochemical parameters. Differential scanning calorimetry and x-ray diffraction studies indicated the amorphized nature of ASPM in lipid matrix. In vitro drug release study indicated the sustained release of drug from NLCs. ASPM-NLCs showed greater permeability across Caco2 cells and everted rat ileum. ASPM-NLCs showed greater cellular uptake, superior preclinical oral bioavailability and higher efficacy in reducing the L- OPA-carbidopa-induced locomotor count compared with plain drug. Conclusion: ASPM-NLCs were successfully developed that showed enhanced performance both in vitro and in vivo

3.  Mehta, Chetan Hasmukh and Reema, Narayan and Nayak, Usha Y (2019) 

Computational modeling for formulation design.

Drug Discovery Today, 24 (3). pp. 781-788. ISSN 1359-6446

Formulation design is an important phase in the drug development process. However, this process at an experimental level requires exhaustive experimental work. Excipient selection, prediction of solubility,encapsulation efficiency, release patterns, drug absorption, stability, and mechanism of nanoparticle formation are some of the essential steps in formulation design. The use of various computational tools,including quantitative structure–activity relationships (QSARs), molecular modeling, molecular mechanics, discrete element modeling, finite element method, computational fluid dynamics, and physiologically based pharmacokinetics (PBPK) modeling, help in the identification of drug product inadequacies and to recommend avenues for understanding complex formulation design in less time with lower investment. Here, we focus on computational modeling tools used in formulation design and its applications.

4.  Vamshi, Krishna T (2019) 

Evaluation of penetration process into young and elderly skin using confocal Raman spectroscopy.

Vibrational Spectroscopy, 100 (1). pp. 123-130. ISSN 0924-2031

Non-invasive in vivo human studies have always been of great importance owing to their advantages over the invasive studies. This research work also presents such a non-invasive study dealing with the penetration of two vitamin derivatives: retinyl acetate and alpha-tocopheryl acetate, into the stratum corneum of two groups of study participants categorized based on age as young (average age of 24.1 ± 3.3 years old) and elderly groups (average age of 68 ± 5.8 years old). According to the increase of age it is expected that intrinsic alterations may occur in the stratum corneum. Understanding these alterations is relevant to the knowledge of the differences between stratum corneum as a function of age-dependence thus, leading to the development of new specific products for ageing. The penetration of these derivatives was semi-quantitatively analyzed using confocal Raman spectroscopic technique with 3510 Skin Composition Analyzer (River Diagnostics, Rotterdam, The Netherlands) with a diode laser of 785 nm wavelength and 26 mW power. The analysis was done in the extended fingerprint region (1800 - 400 cm−1 ) for the stratum corneum region of the skin considering the depths from surface up to 24 μm. The results of this study clearly indicated that these two vitamin derivatives penetrated significantly into the stratum corneum of both study groups and their penetration was mainly affected by the composition of SC and their physico-chemical properties. The penetration profile of the alpha-tocopheryl acetate displays significant statistical difference (p < 0.05) between groups from surface up to 12 μm.

5.  Shreya, AB and Raut, Sushil Yadaorao and Managuli, Renuka Suresh and Udupa, N and Mutalik, Srinivas (2019) 

Active targeting of drugs and bioactive molecules via oral administration by ligand-conjugated lipidic nanocarriers: Recent advances.

AAPS PharmSciTech, 20 (15). pp. 1-12. ISSN 1530-9932

The oral route is the most widely accepted and commonly used route for administration. However, this route may not be suitable for certain drug candidates which suffer from the problem of low aqueous solubility and gastrointestinal absorption and extensive first-pass effect. Nanotechnology-based approaches can be taken up as remedies to overcome the disadvantages associated with the oral route. Among the various nanocarriers, lipidic nanocarriers are widely used for oral delivery of bioactive molecules owing to their several advantages. Active targeting of bioactive molecules via lipidic nanocarriers has also been widely attempted to improve oral bioavailability and to avoid first-pass effect. This active targeting approach involves the use of ligands grafted or conjugated onto a nanocarrier that is specific to the receptors. Active targeting increases the therapeutic efficacy as well as reduces the toxic side effects of the drug or bioactive molecules. This review mainly focuses on the challenges involved in the oral delivery of drugs and its approaches to overcome the challenges using nanotechnology, specifically focusing on lipidic nanocarriers like liposomes, solid lipid nanoparticles, and nanostructured lipid carriers and active targeting of drug molecules by making use of ligand-conjugated lipidic nanocarriers.

 

2018

  1. Siddarth Jadeja, Girish Pai, Krishnamurthy Bhat, MudduKrishna BS. President’s emergency plan for AIDS relief. Sys. Rev. Pharm, 2018. 9(1): 6-9.
  2. Swapnil Kumar, Reema Narayan, Vasif Ahammed, Yogendra Nayak, Anup Naha, Usha Y Nayak. Development of ritonavir solid lipid nanoparticles by Box Behnken design for intestinal lyphatic targeting. J of Drug Delivery Science and Technology, 2018. 44: 181-189. (IF 1.194)
  3. Mukundkumar Rameshbhai Hirpara, Jyothsna Manikkath, K. Sivakumar, Renuka S. Managuli  Karthik Gourishetti, Nandakumar Krishnadas, Rekha R. Shenoy, Belle Jayaprakash, Chamallamudi Mallikarjuna Rao, Srinivas Mutalik. Long circulating PEGylated-chitosan nanoparticles of Rosuvastatin calcium: Development and in vitro and in vivo evaluations. Int. J. Biological Macromolecules, 2018. 107: 2190-2200. (IF 3.671).
  4. Haritima Joshi, Aswathi R. Hegde, Pallavi K. Shetty, Hemanth Gollavilli, Renuka S. Managuli, Guruprasad Kalthur, Srinivas Mutalik. Sunscreen creams containing naringenin nanoparticles: Formulation development and in vitro and in vivo evaluations. Photodermatology, Photoimmunology & Photomedicine, 2018. 34(1): 69-81.  (IF 2.662).
  5. Vamshi Krishna T, Thiago de Oliveira Mendes and Airton Abrahao Martin. In Vivo human skin study of sunscreens by Confocal Raman Spectroscopy. AAPS PharmSciTech, 2018. 19(2) 753-760. (IF 2.451).
  6. Lalit Kumar, Ranamalla S Reddy, Chinna R Pydi, Vamshi Krishna T, Muddukrishna B and Girish Pai K. A Research on market complaint product: Detailed investigation and a report on broken film coated tablet inside an intact blister pack. Latin Am J Pharm, 2018. 37(2): 363-367. (IF 0.298).
  7. Vasanti Suvarna, Siddhata Thorat, Usha Nayak, Atul Sherje, Manikanta Murahari. Host-guest interaction study of Efavirenz with hydroxypropyl-β-cyclodextrin and L-arginine by computational simulation studies: Preparation and characterization of supramolecular complexes. J Molecular Liquids, 2018. 259: 55-64. (IF 3.648).
  8. Rupa Tunge, Mahalaxmi Rathnanand, Gasper Fernandes, Kartik Sharma. A Review on approaches used to overcome problems associated with conventional dosage forms of H2 Antagonist, 2018. 10(4): 1-8.
  9. Muddukrishna B S, Vamshi Krishna T, Lalit Kumar, Girish Pai K. Importance of Competency level and its assessment in Pharmaceutical Industry, Res. J Pharm. Tech. 2018. 11(1): 139-142.
  10. Saran Gopalakrishnan, Eshwari Dathathri, Goutam Thakur, K.B. Koteshwara. Development of Polyvinlyl Alcohol-ChitosanHydroxypropylmethylcellulose Based Composite Films for Controlled Drug Delivery, Trends Biomater. Artif. Organs. 2017. 31(3), 97-101.
  11. Gasper J. Fernandes,Lalit Kumar Kartik Sharma Rupa Tunge and Mahalaxmi Rathnanand. A Review on Solubility Enhancement of Carvedilol—a BCS Class II Drug, Journal of Pharmaceutical Innovation. 2018. 14(1), 1-16. (IF 2.234).
  12. Lalit KUMAR & Ruchi VERMA. Design, Drug Likeness and Molecular Docking of Novel Molecules for Anti- Tubercular Activity. Lat. Am. J. Pharm. 2018. 37 (4): 759-67 (IF 0.298).
  13. Ekta NAIK, Divya HARMALKAR, Soraiya GODINHO, Lalit KUMAR, Shaila LEWIS& Rupesh K. SHIRODKAR. Liquisolid Technique Using Tween 80 and Sodium Starch Glycolate to Enhance the Solubility and Dissolution Rate of an Antihypertensive Drug, Lat. Am. J. Pharm. 2018. 37 (4): 717-26 (IF 0.298).
  14. Pam Wen Lung1 & Vamshi Krishna Tippavajhala2 & Thiago de Oliveira Mendes3 & Claudio A. Téllez-Soto, Pam Wen Lung, Vamshi Krishna Tippavajhala, Thiago de Oliveira Mendes, Claudio A. Téllez-Soto,Desirée Cigaran Schuck, Carla Abdo Brohem,  Marcio Lorencini,  Airton Abrahão Martin. In vivo study of dermal collagen of striae distensae by confocal Raman spectroscopy, Lasers in Medical Science. 2018. 33:609–617 (IF 2.299).
  15. Harsha Vardhani Kondepati, Girish Pai Kulyadi, Vamshi Krishna Tippavajhala. A Review on In Situ gel forming ophthalmic drug delivery systems, Research J. Pharm. and Tech, 2018. 11(1):380-386.
  16. Abhishek Sunil Dhoot, Anup Naha, Juhi Priya, Neha Xalxo. Phase Diagrams for Three Component Mixtures in Pharmaceuticals and its Applications. J. Young Pharmacists, 2018. 10(2): 132-137.
  17. Juhi Priya, Anup Naha, Abhishek Sunil Dhoot, Neha Xalxo. A Review on Polymeric Nanoparticles: A Promising Novel Drug Delivery System. J Global Pharma Tech, 2018. 10(4): 10-17.
  18. Aswathi R. Hegde, Renuka S. Managuli, Anup Naha, Kunnatur B. Koteshwara, Meka Sreenivasa Reddy and Srinivas Mutalik. Full Factorial Experimental Design for Development and Validation of a RP-HPLC Method for Estimation of Letrozole in Nanoformulations. Current Pharmaceutical Analysis, 2018. 14(3): 320-330 (IF 0.75).
  19. Abhishek Chandra, M. Vivek Ghate, K. S. Aithal, Shaila A. Lewis. In silico prediction coupled with in vitro experiments and absorption modeling to study the inclusion complex of telmisartan with modified beta-cyclodextrin. Journal of Inclusion Phenomena and Macrocyclic Chemistry, 2018. 91:47–60 (IF 1.253).
  20. Gururaj C. Aithal, Usha Yogendra Nayak, Chetan Mehta, Reema Narayan, Pratibha Gopalkrishna, Sudharsan Pandiyan and Sanjay Garg. Localized In Situ Nanoemulgel Drug Delivery System of Quercetin for Periodontitis: Development and Computational Simulations. Molecules, 2018. 23(1363): 1-15 (IF 2.861).
  21. Zenab Attari, Lalit Kumar, C. Mallikarjuna Rao, and K. B. Koteshwara. Reversed-Phase HPLC Method for Determination of Temozolomide In rat plasma and Brain: Simple, Sensitive and Robust Method. Pharmaceutical Chemistry Journal, 2018. 52(3): 266-270(IF 0.445).
  22. Shaik Baji, Aswathi R. Hegde, Mrugank Kulkarni, Sushil Yadaorao Raut, Jyothsna Manikkath, Meka Sreenivasa Reddy, Srinivas Mutalik. Skin permeation of gemcitabine hydrochloride by passive diffusion, iontophoresis and sonophoresis: In vitro and in vivo evaluations. J Drug Del Tech, 2018. 47(6): 49-54 (IF 2.297).
  23. Shaik Naaz, Vamshi Krishna Tippavajhala. Hot-Melt Extrusion Technology in the Emerging Pharma Field. Res. J Pharm. Tech, 2018. 11(4): 1619-1623.
  24. Girish Pai K, Tirumala Chetty Sudheer Kumar, Durga Prasad Parida, Alex Joseph. A Review on Manufacturing Operations of Solid Dosage Forms: Design and Implementation on GMP Systems in the Manufacturing Facility. J Global Pharm Tech, 2018. 10(06): 1-10.
  25. Abishek Wadhwa, Vashish Mathura, Shaila Angela Lewis. Emerging Novel Nano-pharmaceuticals for Drug Delivery. Asian J Pharmaceutical and Clinical Research, 2018. 11(7): 35-42.
  26. Reema Narayan, Usha Y. Nayak, Ashok M. Raichur and Sanjay Garg. Mesoporous Silica Nanoparticles: A Comprehensive Review on Synthesis and Recent Advances. Pharmaceutics, 2018. 10(118): 1-50 (IF 3.746).
  27. Arun K. Kodoth, Vivek M. Ghate, Shaila A. Lewis, Vishalakshi Badalamoole. Application of pectin zinc oxide hybrid nanocomposite in the delivery of a hydrophilic drug and a study of its isotherm, kinetics and release mechanism. Int. J. Biol. Macro, 2018. 115: 418-430 (IF 3.909).
  28. Jessica Fernandesa, M. Vivek Ghatea, Sanchari Basu Mallikb, Shaila A. Lewis. Amino acid conjugated chitosan nanoparticles for the brain targeting of a model dipeptidyl peptidase-4 inhibitor. Int. J Pharm, 2018. 547:563-571 (IF 3.862).
  29. Abhishek Chandra, Ankita D. Chondkar, Rupesh Shirodkar, Shaila A. Lewis. Rapidly dissolving lacidipine nanoparticle strips for transbuccal administration. J. Drug Del. Sci. Tech, 2018. 47: 259–267 (IF 2.297).
  30. Renuka Suresh Managuli, Sushil Yadaorao Raut, Meka Sreenivasa Reddy & Srinivas Mutalik. Targeting the intestinal lymphatic system: a versatile path for enhanced oral bioavailability of drugs. Expert Opinion on Drug Delivery, 2018. 15(8):n787-804 (IF 5.553).

 

CIF = 46.026.

 

2017

  1. Sharnya Sankaran Kartha, Girish Pai K, Krishnamurthy Bhat and Muddukrishna BS. Switching Drugs from Rx to OTC status – A regulatory Prespective. J Young Pharm, 2017. 9(1): 3-7.
  2. Naya Sree KS, Ruchi Verma, Girish Pai K and Lalit Kumar. A quality by design approach on polymeric nanocarrier delivery of gefitinib: formulation, in vitro and invivo characterization. Int. J. Nanomed, 20117. 12: 15-18. (IF 4.32).
  3. Paula R Chellini, Thiago O Mendes, Pedro HC Franco, Brenda LS Porto, Vamshi K Tippavajhala, Isabela C Cesar, Marcone AL Oliveira and Gerson A Pianetti. Simultaneous determination of rifampicin, isoniazid, pyrazinamide and ethambutol in 4-FDC tablet by Raman spectroscopy associated to chemometric approach. Vibrational Sepctrosocpy, 2017. 90: 14-20. (IF 1.682).
  4. Gopal Venkatesh Shavi, Usya Yogendra Nayak, Meka Sreenivasa Reddy, Kishore Ginjupalli, Praful Balvant Deshpande, Ranjith Kumar Averineni, Nayanabhirama Udupa, Satya Sai Sadhu, Cyril Danilenkoff and Ramesh Raghavendra. A novel long-acting biodegradable depot formulation of anastrazole for breast cancer therapy. Material Science and Eng. C, 2017. 75: 535-544. (IF 3.42).
  5. Jyothsna Manikkath, Aswathi R Hegde, Guruprasad Kalthur, Harendra S Parekh, Srinivas Mutalik. Influence of peptide dendrimers and sonophoresis on the transdermal delivery of ketoprofen. Int. J Pharm, 2017. 521: 110-119. (IF 3.994).
  6. Muthukumar Amirthalingam, Narayanan Kasinathan, Arul Amuthan, Srinivas Mutali, M Sreenivasa Reddy and Udupa Nayanabhirama. Bioactive PLGA-curcumin microparticle-embedded chitosan scaffold: invitro and in vivo evaluation. Artifical Cells, Nanomedicine, and Biotechnology, 2017. 45(2): 233-241. (IF 2.024).
  7. Kiran S Avadhani, Jyothsna Mannikkath, Mradul Tiwari, Misra Chandrasekhar, Ashok Godavarthi, Shimoga M Vidya, Raghu C Hariharapura, Guruprasad Kalthur, Nayanabhirama Udupa and Srinivas Mutalik. Skin delivery of epigallocatechin-3gallate (EGCG) and hyaluronic acid loaded nano-transfersomes for antioxidant and anti-aging effects in UV radiation induced skin damage. Drug Delivery, 2017. 24(1): 61-74. (IF 4.843).
  8. Jino Elsa Thomas, Usha Y Nayak, Jagadish PC, Koteshwara KB. Design and characterization of Valsartan c0-Crystals to improve its Aqueous Solubility and Dissolution Behavior. Research J. Pharm and Tech, 2017. 10(1): 26-30.
  9. Ashish Hanumantrao Chamle, Nazare Lobo John Shane, Girish Pai, Aravind Pai, Muddukrishna Badamane Sathyanarayana. RP-HPLC method development and validation for the estimation of Methylcobalamin in Bulk. J of Global Pharma Tech, 2017. 03(9): 06-12.
  10. M Sreenivasa Reddy, L Kumar, Z attari and R Verma. Statistical optimization of extraction process for the quantification of Valsartan in rabbit plasma by a HPLC method. Indian Journal of Pharm. Sci, 2017. 79(1): 16-28. (IF 0.762).
  11. Karthik Aithal, Aravind Pai, Girish Pai and Muddukrishna BS. Preparation, solid state characterization of Etraverine Co-Crystals with improved solubility. Latin Am. J. Pharm., 2017. 36(5): 972-979. (IF 0.329).
  12. Atul P Sherje, Vaidehi Kulkarni, Manikanta Murahari, Usha Y Nayak, Pritesh Bhat, Vasanti Suvarna and Bhushan Dravyakar. Inclusion complexation of Etodolac with hydroxypropyl -beta-cyclodextrin and Auxiliary agents: Formulation, characterization and molecular modeling studies. Mol. Pharmaceutics., 2017. 14: 1231-1242. (IF 4.342).
  13. Deepthi Soma, Zenab Attari, Meka Sreenivasa Reddy, Atmakuri Damodaram, Kunnatur Balasundara Gupta Koteshwara. Solid lipid nanoparticles of irbesartan: preparation, characterization, optimization and pharmacokinetic studies. Braz. J. Pharm. Sci., 2017. 53(1): 1-10. (IF 0.485).
  14. Aswathi R Hegde, Prarthana V Rewatkar, Jyothsna Manikkath, Karnaker Tupally, Harendra S Parekh, Srinivas Mutalik. Peptide dendrimer-conjugates of ketoprofen: Synthesis and ex vivo and in vivo evaluations of passive diffusion, sonophoresis and iontophoresis for skin delivery. Eu. J. Pharm. Sci., 2017. 102: 237-249. (IF 3.773).
  15. Nazare Lobo John Shane, Ashish Hanumantrao Chamle, Vasantharaju, Aravind pai, Girish Pai, Muddukrishna BS. Fabrication and solid state characterization of Ticagrelor co-crystals with improved solubility and dissolution. Int. J Pharm Qual. Assu., 2017. 18(1):1-8.
  16. Aravind Pai, Mahalaxmi Rathnanand and Madhu Kumar. Stability enhancement of proton pump inhibitor in stomach: formulation and in vitro evaluation of stabilized proton pump inhibitor. Asian J Pharm and Clin Res., 2017. 10(5): 88-92.
  17. Naya Sree KS, Girish Pai K, Ruchi Verma, Padiya Ananthakrishna and Lalit Kumar. Validation of HPLC method for quantitative determination of Gefitinib in polymeric nanoformulation. Pharm. Chem J., 2017. 51(2): 159-163. (IF 0.461).
  18. Jay M Jetani and Girish Pai K. A Comparative Review of the USFDA Guidelines on Process Validation Focusing on the Importance of Quality by Design (QbD). Research J. Pharm. and Tech., 2017. 10(4): 1257-1260.
  19. Ahammed V, Narayan R, Paul J, Nayak Y, Roy B, Shavi GV, Nayak UY. Development and in vivo evaluation of functionalized ritonavir proliposomes for lymphatic targeting. Life Sciences, 2017. 183:11-20. (IF 2.936)
  20. Reema Narayan, Abhyuday Pednekar, Dipshikha Bhuyan, Chaitra Gowda, Koteshwara KB, Usha Yogendra Nayak. A top-down technique to improve the solubility and bioavailability of aceclofenac: In vitro and In vivo studies. Int. J. Nanomed, 2017. 12: In Press (IF 4.30).

 

2016

  1. Lalit Kumar, Meka S. Reddy, Ruchi Verma and KB KOTESHWARA. Selection of Cryoprotective Agent for Freeze Drying of Valsartan Solid Lipid Nanoparticles, Lat. Am J. Pharm. 2016, 35(2), 284-290.
  2. Sunetra Kalvakuntla, Mangesh Deshpande, Zenab Attari, Koteshwara KB. Preparation and characterization of Nanosuspension of Aprepitant by H96 Process. Adv. Pharm. Bull. 2016, 6(1), 83-90.
  3. Damodaram Atmakuri, Zenab Attari, Girish Pai, Koteshwara KB. Enteric coated capsules of eprosartan mesylate withpH-modifier for augmenting the dissolution. Int. J. Res. Pharm. Sci. 2016, 7(1), 34-38.
  4. Gopal Venkatesh Shavi, Meka Sreenivasa Reddy, Ramesh Raghavendra, Usha Yogendra Nayak, Averineni Ranjith Kumar, Praful Balavant Deshpande, Nayanabhirama Udupa, Gautam Behl, Vivek Dave & Kriti Kushwaha. PEGylated liposomes of anastrozole for long-term treatment of breast cancer: in vitro and in vivo evaluation. J Liposome Res. 2016, 26(1): 28–46.
  5. Zenab Attari, Lalit Kumar, C Mallikarjuna Rao, KB Koteshwara. Validation of sensitive and robust reversed phase-HPLC method for determination of temozolomide. Lat. Am. J. Pharm. 2016, 35(1): 967-971.
  6. Rupesh K Shirodkar, Lalit Kumar, Chandrashekar Misra, Renuka Managuli, Zenab Attari, Srinivas Mutalik and Shaila A Lewis.  Development and validation of a reverse phase HPLC method for the estimation of Lacidipine in rat plasma:application to preclinical pharmacokinetics. Lat. Am. J Pharm. 2016, 35(1): 124-129.
  7. Zenab Attari, Amita Bhandari, PC Jagadish and Shaila Lewis. Enhanced ex vivo intestinal absorption of olmesartan medoxomil nanosuspension: preparation by combinative technology. Saudi Pharm J. 2016, 24: 57-63.
  8. T Darekar, K S Aithal, R Shirdkar, L Kumar, Z attari and S. Lewis. Characterization and in vivo evaluation of lacidipine inclusion complexes with β-cyclodextrin and its derivatives. J Incl Phenom Macrocyl Chem. 2016, 84: 225-235.
  9. Ushasree Medikonduri, Zenab Attari, Krishnamurthy Bhat and Shaila Lewis.  Acyclovir entrapped N-trimethyl chitosan nanoparticles for oral bioavailability enhancement. Cur. Nanosci. 2016, 12: 378-385.
  10. Amita Bhandari, Zenab Attari, Prashant Musmade and Shaila Lewis. Enhanced intestinal permeability of Quercetin coated Feodipine beads in Wistar rats. Formulation and Ex Vivo study. Lat. Am. J Pharm. 2016, 35(3): 558-563.
  11. Usha Y Nayak, Mutalik S and Udupa N. An overview of novel and controlled drug delivery systems. Indian Drugs. 2016, 53(5): 05-12.
  12. Srinivas Mutalik, Neelam A Suthar, Renuka S Managuli, Pallavi K Shetty, Kiran Avadhani, Guruprasad Kalthur, Raghavendra V Kulkarni and Ranjeny Thomas. Development and performance evaluation of novel nanoparticles of a grafted copolymer loaded with curcumin. Int. J. Biol. Macro. 2016, 86(5): 709-720.
  13. Paridhi Bhargava, Swati Pralhad, Sshita S Uppoor, Dilip G Nayak, Srinivas Mutalik and Pallavi Shetty. Evaluation of antibacterial, antioxidant, anti-inflammatory and cytotoxicity potential of mouth rinse containing piper betel essential oil. Int. J. Res. Ayurveda Pharm. 2016, 7(2): 217-221.
  14. Renuka S Managuli, Lalit Kumar, Ankita D Chonkar, Rupesh K Shirodkar, Shaila Lewis, Kunnatur B Koteshwara, Meka Sreenivasa Reddy and Srinivas Mutalik. Development and validation of a stability-indicating RP-HPLC method by a statistical optimization process for the quantification of Asenapine maleate in lipidic nanoformulations. J. Chrom Sci. 2016, 5:1-11.
  15. Pradeepa, SM Vidya, Srinivas Mutalik, K Udaya Bhat, Prashant Huligol and Kiran Avadhani. Preparation of gold nanoparticles by novel bacterial exopolysaccharide for antibiotic delivery. Life Sciences. 2016, 153(6): 171-179.
  16. Yogendra Nayak, Kiran Avadhani, Srinivas Mutalik and Usha Y Nayak. Lymphatic Delivery of Anti-HIV Drug nanoparticles. Recent Patents in Nanotechnology. 2016, 10:116-127. 
  17. Phani Krishna Kondamudi, Phani Prashanth Tirumalasetty, Rajkumar Malayandi, Srinivas Mutalik and Raviraj Pillai. Lidocaine transdermal patch: Pharmacokinetic modeling and in vitro-in vivo correlation (IVIVC). AAPS PharmSciTech. 2016. 17(3): 588-596. 
  18. Saurabh Dubey, Kiran Avadhani, Srinivas Mutalik, Sangeetha Madambithara Sivadasan, Biswajit Maiti, Joydeb Paul, Shivani Kallappa Girisha, Moleyur Nagarajappa Venugopal, Stephen Mutoloki, oystein Evensen, Indrani Karunasagar and hetron mweemba Munang’andu. Aeromonas hydrophila OmpW PLGA Nanoparticle oral vaccine shows a dose-dependent protective immunity in Rohu (Labeo rohita). Vaccines. 2016. 4(21): 1-11.
  19. A B Shreya, Renuka S Managuli, Jyothsna Menon, Lavanya Kondapalli, Aswathi R Hegde, Kiran Avadhani, Pallavi K Shetty, Muthukumar Amirthalingam, Guruprasad Kalthur and Srinivas Mutalik. Nano-transfersomal formulations for transdermal delivery of asenapine maleate: in vitro and in vivo performance evaluation. J. Liposome Res. 2016. June 2016: 221-232. 
  20. Pradeepa, Akshay D Shetty, Koshi Matthews, Aswathi R Hegde, B Akshatha, Alvita Betilla Mathias, Srinivas Mutalik and S M Vidya. Multidrug resistant pathogenic bacterial biofilm inhibition by Lactobacillus plantarum exopolysaccharide. Bioactive Carbohydrates and Dietary Fibre. 2016. (1): 07-14.
  21. Rashmi Bopanna, Raghavendra V Kulkarni, G Krishna Mohan, Srinivas Mutalik and Tejraj M Aminabhavi. Invitro and invivo assessment of novel pH-sensitive interpenetrating polymer networks of a graft copolymer for gastro-protective delivery of ketoprofen. RSC.Adv. 2016. 6: 64344-64356. 
  22. Ankita D Chonkar J Venkat Rao, Renuka S Managuli, Srinivas Mutalik, Swapnil Dengale, Prateek Jain and N Udupa. Development of fast dissolving oral films containing lercanidipine HCl nanoparticles in semicrystalline polymeric matrix for enhanced dissolution and ex vivo permeation. Eu. J. Pharm. Biopharm. 2016. 103: 179-191. 
  23. Guruprasad Nayak, Sachin D Honguntikar, Sneha Guruprasad Kalthur, Antony Sylvan D’Souza, Srinivas Mutalik, Manjunath M Setty, Raksha Kalyankumar, Hanumanthappa Krishnamurthy, Guruprasad Kalthur and Satish Kumar Adiga. Ethanolic extract of Moringa Oleifera Lam. leaves protect the pre-pubertal spermatogonial cells from cyclophosphamide-induced damage. Journal of Ethnopharmacology. 2016. 182: 101-109. 
  24. Sakaram Kashyap Khatri, Mahalaxmi Rathnand and Nikhila R. Formulation and evaluation of wound healing activity of linezolid topical preparations on diabetic rats. Int. J. App. Pharmaceu. 2016. 8(3): 30-36.
  25. Siddharth Jadeja, Girish Pai Kulyadi and Muddukrishna BS. Adoption and reasons for withdrawal of ICH Q1F guidelines. J Young Pharm, 2016. 8(4): 500-504.
  26. Girish Pai Kulyadi and Muddukrishna BS. Stability indicating HPTLC determination of Triamcinalone acetoinde in bulk drug and sterile injectable suspension. J Young Pharm. 2016. 8(4): 430-435.
  27. Arpitha Rao, Guruprasad Nayak, Sandhya Kumari, Amratha D Prabhu, Nalini Khandige, Sneha Guruprasad Kalthur, Srinivas Mutalik, Guruprasad Kalthur and Satish Kumar Adiga. Ethambutol induces testicular damage and decreases the sperm functional competence in Swiss albino mice. Environmental Toxicology and Pharmacology, 2016. 47:28-37. 
  28. Praful Balvant Deshpande, Aravind Kumar Gurram, Amruta Deshpande, Gopal Venkatesh Shavi, Prashant Musmade, Karthik Armugam, Ranjith Kumar Averineni, Srinivas Mutalik, Meka Sreenivasa Reddy and Nayanabhirama Udupa. A novel nanoproliposomes of lercanidipine: Development, in vitro and preclinical studies to support its effectiveness in hypertension therapy. Life Sciences, 2016. 162: 12-137. 
  29. Kiran S Avadhani, Muthukumar Amirthalingam, Meka Sreenivasa Reddy, Nayanabhirama Udupa and Srinivas Mutalik. Development and Validation of RP-HPLC method for estimation of Epigallocatechin-3gallate (EGCG) in lipid nanoformulations. Research J Pharm and Tech., 2016. 9(6): 725-730.
  30. Thiago de oliveira Mendes, Liliane Pereira Pinto, Laurita dos Santos, Vamshi Krishna Tippavajhala, Claudio Alberto Telle Soto and Airton Abrahao Martin. Stastical strategies to reveal potential vibrational markers for in vivo analysis by confocal Raman spectroscopy. J Biomed. Optics, 2016. 21(7): 075010-1 to 075010-10.
 

ARCHIVES

 

2010 - 2015