QbD enabled Process Variable Study to Develop Sustained Release Chitosan-Alginate Embedded Delivery System for Improved Patient Compliance

Autores

  • Vijay Sharma Pharmacy Academy, IFTM University, Moradabad, India https://orcid.org/0000-0003-1898-0476
  • Lalit Singh Future Group of Institutions, Bareilly, india
  • Navneet Verma Pharmacy Academy, IFTM University, Moradabad, India

DOI:

https://doi.org/10.1590/s2175-97902021000319803%20%20

Palavras-chave:

Quality target product profile, Critical quality attributes, Critical material attributes

Resumo

The current investigation entail systematic Quality by Design (QbD)-enabled approach for the development of Sustained released embedded drug delivery systems of L-Arginine employing ionic gelation technique to attain improved patient compliance. Hence, in this QbD enabled systematic approach; quality target product profile (QTTP) was defined and critical quality attributes (CQAs) were identified. Further the risk assessment studies were undertaken through Ishikawa fish bone diagram to locate the critical material attributes (CMAs) and/or critical process parameters (CPPs) for the formulation of beads that may affect CQAs of drug product. A face centered central composite design (CCD) for two factors at three levels each with α =1 was employed for the optimization process to checkout the impact of concentration of sodium alginate and concentration of chitosan as CMAs which wereprior identified from risk assessment study and further evaluated for CQAs viz. bead size, swelling index and percent drug entrapment. The optimum formulation was embarked upon by using mathematical model being developed yielding desired CQAs. Thereby chitosan coated calcium-alginate delivery system was successfully developed by strategically employing QbD approach.In a nutshell, the presentinvestigation reports the successful development of optimized chitosan coated alginate beads employing QbD approach which can serve as a platform for other drugs too.

Downloads

Os dados de download ainda não estão disponíveis.

Referências

Abdelbary A, El-Gazayerly ON, El-Gendy NA, Ali AA. Floating tablet of trimetazidine dihydrochloride: An approach for extended release with zero-order kinetics. AAPS PharmSciTech. 2010;11(3):1058-1067.

Aral C, Akbuğa J. Alternative approach to the preparation of chitosan beads. Int J Pharm Sci. 1998;168(1):9-15.

Chien YW. Concepts and system design foe rate-controlled drug delivery in novel drug delivery systems 2nd ed. New York:Marcel Dekker Inc; 1992. P. 50:1-42.

Dorozynski P, Jachowicz R, Kulinowski P, Kwiecinski S, Szybinski K, Skorka T. The macromolecular polymers for the preparation of hydrodynamically balanced systems--methods of evaluation. Drug Dev Ind Pharm. 2004;30(9):947-57.

Douglas KL, Tabriziani M. Effect of experimental parameters on the formation of alginate-chitosan nanoparticles and evaluation of their potential application as DNA carrier. J Biomater Sci Polym. 2005;16(1):43-56.

Fahmy R, Kona R, Dandu R. Quality by design I: application of failure mode effect analysis (FMEA) and Plackett- Burman design of experiments in the identification of ‘‘main factors’’ in the formulation and process design space for roller-compacted ciprofloxacin hydrochloride immediate-release tablets. AAPS PharmSciTech . 2012;13(4):1243-54.

George M, Abraham TE. Polyionic hydrocolloids for the intestinal delivery of protein drugs: alginate and chitosan-a review. J Cont Rel. 2006;114(1):1-14.

George P, Nikolaos B. Swelling studies and in vitro release of verapamil from calcium alginate and calcium alginate-chitosan beads. Int J Pharm. 2006;323(1-2):34-42.

Gombotz WR, Wee SF. Protein release from alginate matrices. Adv Drug Deliv Rev. 1998;31(3):267-85.

Hedin SG.A method to isolate the lysine, together with some remarks about the lysatinin. Physiol Chem. 1895;21:297-305.

Jantzen GM, Robinson JR, Lee VHL.Design and fabrication of oral controlled release drug delivery systems. In: Banker GS, Rhodes CT, editors. Modern Pharmaceutics. 3rd ed. New York, Marcel Dekker Inc. 1996: p. 123-127.

Krebs HA, Henseleit H. Investigations on the formation of urea in Tierkoerper. Physiol Chem.1932;210:33-66.

Lalit S, Arun N, Vijay S, Saurabh S. Formulation bydesign: understanding the formulation variables and optimization of glipizide buoyant bioadhesive beads by central composite design. Malay J Pharm Sci. 2014;12(1):1-18.

Majetin V, Kumar R. Nano and microparicle as controlled drug delivery. J Pharm Pharmaceut Sci. 2000;3(2):234-58.

Marsh, JR,Weiss PJ. Solubility of antibiotics in twenty six solvents. J Ass Off Anal Chem.1967;50(8):437-462.

Murrell GA, Szabo C, Hannafin JA, Jang D, Doilan MM, Deng XH, et al. Modulation of tendon healing by nitric oxide. Inflamm Res.1997;46(1):19-27.

Patel YL, Sher P, Pawar AP. The effect of drug concentration and curing time on processing and properties of calcium alginate beads containing Metronidazole by response surface methodology. AAPS Pharmaceutical Science and Technology. 2006;7(4):86.

Prabhakara P, Harish NM, Gulzar AM, Brijesh Y, Narayana CR, Satyanarayana D.Formulation and in-vitro evaluation of gastric oral floating tablets of glipizide. Ind J Pharm Edu Res. 2008;42(2):174-183.

Rodríguez PC, Ochoa AC. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol Rev. 2008;222:180-191.

Sangeetha S, Sakthisaravanan V, Komala M, Harish G, Sivakumar. Design and evaluation of gastroretentive beads of theophylline by ionotropic gelation. Inter J Pharmacy Pharm Sci. 2010;2(3):99-101.

Sato Y, Kawashima Y, Tekuchi N. Physicochemical properties to determine the buoyancy of hollow microspheres (microballoons) prepared by the emulsion solvent diffusion method. Eur J Phram Biopharm. 2003;55(3):297-304.

Schulze E, Steiger E, Das U. Arginin. Physiol Chem . 1886;11:43-65.

Schwedhelm E, Maas R, Freese R, Jung D, Lukacs Z, Jambrecina A, et al. Pharmacokinetic and pharmacodynamic properties of oral L-citrulline and L-arginine: impact on nitric oxide metabolism. Br J Clin Pharmacol.2008;65(1):51-59.

Şenel S, Hıncal AA. Drug permeation enhencement via buccal route: possibilities and limitations. J Contr Rel. 2001;72(1-3):133-44.

Sharma V, Singh L, Verma N. Formulation development of Sustained release alginate beads of amino acid. European Journal of Biomedical and Pharmaceutical sciences. 2017;4(9):653-659.

Sherina VM, Santhi K, Sajeeth CIM. Formulation and evaluation of sodium alginate beads as a carrier for the controlled release of nifedipine. Int J Pharm Chem Sci. 2012;1(2):699-710.

Takahashi T, Takayama K, Machida Y, Nagai T. Characteristics of polyion complexes of chitosan with sodium alginate and sodium polyacrylate. Int J Pharm. 1990;61(1-2):35-41.

Tonnesen HH, Karlsen J. Alginate in drug delivery. Drug Dev Ind Pharm . 2002;28(6):621-30.

Xu Y, Zhan C, Fan L, Wang L, Zheng H. Preparation of dual crosslinked alginate-chitosan blend gel beads and in vitro controlled release in oral site-specific drug delivery system. Int J Pharm .2007;336(2):329-337.

Yotsuyanagi T,Ohkubo T,Ohhashi T, Ikeda K. Calcium-induced gelatin of alginic acid and pH-sensitive reswelling of dried gels. Chem Pharm Bull. 1987;35(4):1555-1563.

Downloads

Publicado

2022-11-23

Edição

Seção

Original Article

Como Citar

QbD enabled Process Variable Study to Develop Sustained Release Chitosan-Alginate Embedded Delivery System for Improved Patient Compliance. (2022). Brazilian Journal of Pharmaceutical Sciences, 58. https://doi.org/10.1590/s2175-97902021000319803