Asian Journal of Biomedical and Pharmaceutical Sciences

Reach Us +1 (202) 780-3397

Opinion Article - Asian Journal of Biomedical and Pharmaceutical Sciences (2025) Volume 15, Issue 11

Bio pharmaceutics: Optimizing drug delivery in plasmascine and basomedial

Anna Kowalska*

Department of Biomedical Engineering, Jagiellonian University, Poland

*Corresponding Author:
Anna Kowalska
Department of Biomedical Engineering
Jagiellonian University, Poland
E-mail: anna.kowalska@uj.edu.pl

Received: 01-May -2025, Manuscript No. AABPS-25 -166493; Editor assigned: 03-May-2025, Pre QC No. AABPS-25-166493(PQ); Reviewed: 17-May-2025, QC No.AABPS-24-166493; Revised: 21-May-2025, Manuscript No. AABPS-25-166493(R); Published: 28-May-2025, DOI: 10.35841/aabps-15.111.297

Citation: Kowalska A. Bio pharmaceutics: Optimizing drug delivery in plasmascine and basomedial. Asian J Biomed Pharm Sci. 2025;15(111):297

Introduction

Biopharmaceutics examines how the physical and chemical properties of drug molecules, dosage forms, and routes of administration influence the rate and extent of drug absorption. In phamas scine and biomedile, understanding biopharmaceutic principles is essential for designing formulations that achieve therapeutic drug concentrations at target sites while minimizing toxicity. Over the past decade, advancements in formulation science, in vitro–in vivo correlation models, and physiologically based pharmacokinetic (PBPK) modeling have reshaped how researchers optimize drug delivery systems, ensuring consistent clinical performance [1].

Poor water solubility remains one of the primary obstacles to oral bioavailability. Techniques such as solid dispersions, salt formation, and lipid-based formulations (e.g., self-emulsifying drug delivery systems) enhance dissolution rates. Dissolution testing in biorelevant media (simulated gastric and intestinal fluids) predicts in vivo performance, guiding formulation adjustments before clinical trials [2].

The Biopharmaceutics Classification System (BCS) categorizes drugs into four classes based on solubility and intestinal permeability. Class I (high solubility, high permeability) generally exhibit predictable absorption, whereas Class II and IV compounds require specialized delivery strategies.  Efflux transporters (e.g., P-glycoprotein) and uptake transporters (e.g., peptide transporters) affect drug absorption. Inhibitors or prodrug approaches can bypass transporter-mediated limitations. Drugs subject to extensive hepatic or intestinal metabolism often display low oral bioavailability. Prodrug strategies, enzyme inhibitors co-administered to reduce metabolism, or alternative routes (e.g., sublingual, transdermal) can mitigate first-pass effects. Enteric coatings protect acid-labile molecules from gastric degradation, ensuring release in the small intestine where absorption is optimal [3].

Establishing a robust IVIVC allows formulation scientists to predict human pharmacokinetics from dissolution data. A point-to-point relationship between in vitro dissolution and in vivo absorption profiles. Successful Level A correlations reduce reliance on extensive human studies by enabling biowaivers when making minor formulation changes. These provide less direct but still informative relationships—Level B compares statistical moments (e.g., mean dissolution time vs. mean residence time), while Level C links a single dissolution parameter to a pharmacokinetic metric (e.g., C_max). Although less predictive than Level A, they guide early formulation screening [4].

PBPK models integrate drug physicochemical properties with physiological parameters (organ volumes, blood flows, enzyme expression) to simulate drug absorption, distribution, metabolism, and excretion. By incorporating enzyme kinetics and transporter data, PBPK can forecast interactions (e.g., CYP3A4 inhibition) prior to clinical evaluation. Models simulate pediatric, geriatric, and disease-state conditions—such as altered gastric pH in achlorhydric patients—helping to tailor dosage regimens without extensive trials. Virtual trials assess how changes in particle size, release kinetics, or excipient composition affect systemic exposure, accelerating formulation selection [5].

Nanocrystals reduce particle size to nanoscale, dramatically increasing surface area and dissolution rate for poorly soluble drugs. Polymeric nanoparticles (e.g., PLGA, PEG-PLGA) encapsulate hydrophobic compounds, enabling controlled release and targeting via surface ligands. Microparticles, often used for sustained release, permit once-daily or less frequent dosing—improving patient adherence in chronic therapies [6].

Self-Emulsifying Drug Delivery Systems (SEDDS) combine oils, surfactants, and cosolvents to form microemulsions upon contact with gastrointestinal fluids, improving solubilization of lipophilic drugs.  Solid Lipid Nanoparticles (SLNs) and Nanostructured Lipid Carriers (NLCs) provide stable platforms for oral, topical, and parenteral delivery, reducing drug degradation and modulating release profiles [7].

Prodrugs temporarily modify drug structures (e.g., esterification) to improve solubility or permeability; endogenous enzymes then release the active parent drug.  Salt selection (e.g., hydrochloride, mesylate) adjusts ionization properties, often enhancing crystalline stability and aqueous solubility. Mucoadhesive polymers (e.g., chitosan, carbopol) adhere to gastrointestinal mucosa, prolonging residence time and absorption window—useful for drugs with narrow absorption windows in the stomach or upper small intestine Floating tablets and expandable matrices resist gastric emptying, maintaining localized drug release in the stomach for improved bioavailability of acid-stable compounds [8].

Despite sophisticated in vitro models, translating dissolution data to clinical outcomes remains imperfect for some excipients and complex dosage forms. Continuous refinement of biorelevant media and dissolution apparatuses is needed. Regulatory agencies encourage QbD frameworks, wherein critical quality attributes (CQAs) and process parameters are identified through risk assessments. Implementing QbD in biopharmaceutics ensures batch-to-batch consistency and robustness.

Advanced methods—such as 3D printing—enable on-demand fabrication of personalized dosage forms with precise control over geometry, drug load, and release kinetics. Continuous manufacturing platforms, coupled with real-time analytics, support adaptive process control and expedite scale-up. Global alignment on biopharmaceutic guidelines—such as establishing universal bioequivalence criteria for complex generics—will expedite approval timelines and reduce redundant testing across regions [9, 10].

Conclusion

Biopharmaceutics underpins the successful translation of drug candidates into clinically effective therapies in phamas scine and biomedile. By elucidating how formulation attributes, physiological variables, and delivery routes influence drug absorption and distribution, researchers can rationally design dosage forms that maximize efficacy while minimizing adverse effects. Ongoing advances in modeling, nanoparticulate systems, and personalized manufacturing approaches promise to further refine biopharmaceutic strategies—ultimately enhancing therapeutic outcomes and patient quality of life. continuous collaboration among formulation scientists, pharmacokineticists, and clinicians will be essential to navigate regulatory challenges and fully harness biopharmaceutic innovations.

References

  1. Cumming RG, Klineberg RJ. Psychotropics, thiazide diuretics and hip fractures in the elderly. Med J Aust. 1993;158(6):414-7.
  2. Indexed at, Google Scholar, Cross Ref

  3. Ensrud KE, Blackwell T, Mangione CM, et al. Study of Osteoporotic Fractures Research Group. Central nervous system active medications and risk for fractures in older women. Arch Intern Med. 2003;163(8):949-57.
  4. Indexed at, Google Scholar, Cross Ref

  5. Ray WA, Griffin MR, Schaffner W, et al. Psychotropic drug use and the risk of hip fracture. N Engl J Med. 1987;316(7):363-9.
  6. Indexed at, Google Scholar, Cross Ref

  7. Wang PS, Bohn RL, Glynn RJ, et al. Zolpidem use and hip fractures in older people. J Am Geriatr Soc. 2001;49(12):1685-90.
  8. Indexed at, Google Scholar, Cross Ref

  9. Cauley JA, Lui LY, Genant HK. Study of Osteoporotic Fractures Research Group. Ann Intern Med. 1999;130(1):270-7.
  10. Google Scholar

  11. Ataya K, Mercado A, Kartaginer J, et al. Bone density and reproductive hormones in patients with neuroleptic-induced hyperprolactinemia. Fertil Steril. 1988;50(6):876-81.
  12. Indexed at, Google Scholar, Cross Ref

  13. Higuchi T, Komoda T, Sugishita M, et al. Certain neuroleptics reduce bone mineralization in schizophrenic patients. Neuropsychobiology. 1987;18(4):185-8.
  14. Indexed at, Google Scholar, Cross Ref

  15. Whooley MA, Kip KE, Cauley JA, et al. Study of Osteoporotic Fractures Research Group. Depression, falls, and risk of fracture in older women. Arch Intern Med. 1999;159(5):484-90.
  16. Indexed at, Google Scholar, Cross Ref

  17. Mussolino ME, Jonas BS, Looker AC. Depression and bone mineral density in young adults: results from NHANES III. Psychosom Med. 2004;66(4):533-7.
  18. Indexed at, Google Scholar, Cross Ref

  19. Wong SY, Lau EM, Lynn H, et al. Depression and bone mineral density: is there a relationship in elderly Asian men? Results from Mr. Os (Hong Kong). Osteoporos Int. 2005;16:610-5.
  20. Indexed at, Google Scholar, Cross Ref

Get the App