Decoding of API particle size in reference market product for bio-equivalent and cost effective generic product development

Main Article Content

Jailani.S
C.K.Dhanapal
Noohu Abdulla Khan

Keywords

Decoding, deformulation, reference listed drug, bioequivalence, generic product

Abstract

Decoding or deformulation involves the characterization of the reference listed drug innovator drug where the most important step in deformulation is solid state characterization of API (Active pharmaceutical ingredient). Bioavailability and dissolution rate are directly impacted by API particle-size distribution. It aids in ensuring a bioequivalent formulation, particularly for medications with bioavailability that are dissolution-sensitive. Therefore, the information obtained from the innovator product's API's particle-size distribution is crucial for assuring solubility and bioequivalence. When a generic drug manufacturer experiences multiple Bioequivalence (BE) failures during the development of their product, it can lead to increased developmental costs and ultimately increased product cost the patients. The manufacturer may need to conduct additional studies and testing to optimize the formulation, which can be time-consuming and expensive. Therefore, it's critical to lower the chance of BE failures in order to lower the price of creating generic medicines and the financial burden on patients. Here conventional particle size determination methods are not useful, hot stage microscopy (HSM) is an ideal method , which can be beneficial with microscopic capturing of melting events in the melting point ranges of API and excipients components in a tablet dosage form. In this study, Xarelto 20 mg film-coated tablet was the reference product used and by using hot stage microscopy method, particle size distribution of reference API was found to be 77.4μ and Accordingly, the Rivaroxaban API particles are to be engineered for generic product design and development to have similar particle size distributions based on D90 and D50 values decoded from the reference market product. This can be useful to ensure similar drug performance and efficacy and ultimately favours the bioavailability and would be accessible to low cost as compare to the respective reference product. 

Abstract 208 | PDF Downloads 243

References

1. Munjal B, Koradia V, Boddu SH, Bansal AK. Role of innovator product characterization in generic product development. Excipient applications in formulation design and drug delivery. 2015:521-38.
2. Lionberger RA. FDA critical path initiatives: opportunities for generic drug development. The AAPS journal. 2008 Mar;10:103-9.
3. Curtis SK, Harston SP, Mattson CA. A generic formulaic characterization of the time to reverse engineer the tolerances of a product. InASME International Mechanical Engineering Congress and Exposition 2009 Jan 1 (Vol. 43772, pp. 275-285).
4. Bansal A, Koradia V. The role of reverse engineering in the development of generic formulations. Pharmaceutical Technology. 2005 Aug 2;29(8).
5. York P. Solid-state properties of powders in the formulation and processing of solid dosage forms. International Journal of Pharmaceutics. 1983 Mar 1;14(1):1-28.
6. Martinez MN, Amidon GL. A mechanistic approach to understanding the factors affecting drug absorption: a review of fundamentals. The Journal of Clinical Pharmacology. 2002 Jun;42(6):620-43.
7. Koradia VS, Chawla G, Bansal AK. Comprehensive characterisation of the innovator product: targeting bioequivalent generics. Journal of Generic Medicines. 2005 Jul;2(4):335-46.
8. "Pharmaceutical Solids: A Strategic Approach to Regulatory Considerations," S.R. Byrn et al., Pharm. Res. 12 (7), 945-954 (1995).
9. Gadade DD, Pekamwar SS. Pharmaceutical cocrystals: Regulatory and strategic aspects, design and development. Advanced pharmaceutical bulletin. 2016 Dec;6(4):479.
10. Brittain HG, Bogdanowich SJ, Bugay DE, DeVincentis J, Lewen G, Newman AW. Physical characterization of pharmaceutical solids. Pharmaceutical research. 1991 Aug;8:963-73.
11. Patel P, Ahir K, Patel V, Manani L, Patel C. Drug-Excipient compatibility studies: First step for dosage form development. The Pharma Innovation. 2015 Jul 1;4(5, Part A):14.
12. Izutsu KI, Koide T, Takata N, Ikeda Y, Ono M, Inoue M, Fukami T, Yonemochi E. Characterization and quality control of pharmaceutical cocrystals. Chemical and Pharmaceutical Bulletin. 2016 Oct 1;64(10):1421-30.
13. Šimek M, Grünwaldová V, Kratochvíl B. Hot-stage microscopy for determination of API particles in a formulated tablet. BioMed Research International. 2014 Jul 21;2014.
14. Deore J, Rajput N, Jadav T, Sahu AK, Sengupta P. Hot Stage Microscopy-based Method for Determination of Particle Size in Reverse Engineering: Establishment of a Platform Technology Employing Carvedilol as a Model Drug. Current Analytical Chemistry. 2022 Dec 1;18(10):1117-30.
15. Paliwal R, Mamgain A, Kenwat R, Paliwal SR. Reverse Engineering in Pharmaceutical Product Development. InMicro-and Nanotechnologies-Based Product Development 2021 Sep 6 (pp. 235-242). CRC Press.