Ijraset Journal For Research in Applied Science and Engineering Technology
Authors: Shrikant Subhash Garud , Samadhan Ananda Dhatrak , Thube Rushikesh Sayaji, Amit Ambadas Chaudhari
DOI Link: https://doi.org/10.22214/ijraset.2024.65057
Certificate: View Certificate
The rise of biologics has brought about a major change in modern medicine, offering new ways to treat complex and chronic diseases. Unlike traditional drugs, biologics are created from living cells, making them larger and more intricate. These therapies work by targeting specific parts of the immune system, which helps in treating conditions like cancer, autoimmune disorders, and infectious diseases more effectively. In recent years, biologics have shown great promise in clinical trials, providing better outcomes for patients who haven’t responded well to standard treatments. This review explores how biologics work, their impact on patient care, and emerging trends in this field, such as biosimilars and personalized medicine. Together, these advancements hold potential to improve treatment options and make biologics more accessible.
I. INTRODUCTION
Biologics have dramatically changed the landscape of modern medicine, offering innovative treatments for diseases that were once difficult to manage. Unlike traditional medications, which are often made through chemical processes, biologics are developed from living cells, making them highly complex and precise. They include products like monoclonal antibodies, vaccines, gene therapies, and recombinant proteins, each designed to interact with specific molecules in the body to treat diseases more effectively.
The unique ability of biologics to target specific pathways within the immune system has opened up new treatment possibilities, especially for chronic and complex conditions such as cancer, autoimmune diseases (like rheumatoid arthritis and Crohn’s disease), and certain genetic disorders. These diseases often involve complex interactions within the body, where traditional drugs may fall short due to limitations in targeting specific biological pathways. Biologics address this gap by precisely engaging with targeted cells or proteins, reducing side effects and improving treatment effectiveness.
With biologics, patients who previously had limited treatment options now have access to therapies that can improve their quality of life, slow disease progression, and, in some cases, even offer a cure. The clinical success of biologics has sparked growing interest in this field, prompting significant investment in research and development. As a result, new biologics and biosimilars (essentially lower-cost versions of biologic drugs) are being introduced to the market, broadening access and affordability for patients.
Additionally, the development of biologics aligns with advancements in personalized medicine, which aims to tailor treatments to each patient’s unique genetic and molecular profile. This synergy could potentially enhance the effectiveness of biologic treatments, making them even more precise in addressing the unique needs of individual patients.
In this paper, we will examine the mechanisms behind biologics, the impact they have had on patient care, and the emerging trends that are shaping the future of this field. By understanding these aspects, we can appreciate the potential of biologics to revolutionize healthcare further and address unmet needs in the treatment of challenging diseases.
New technologies enabling the improved delivery of biologics, such as needle-free devices, nanoparticles, and smart nanomaterials, together with the introduction of the concept of personalized medicine resulted in their faster market growth.7,10 One of the most challenging lines is the development of targeted delivery systems for small interfering RNA, which has up to now exhibited superiority under in vitro conditions but failed to achieve targeted as well as intracellular efficient delivery in various in vivo studies.4 Advanced delivery strategies could provide improvements in the targeted delivery of RNA drugs, enabling maximized drug potency while minimizing off-target toxicity and immunogenicity.11 As for all drug therapies, the efficient and targeted delivery of biologics to the desired site of action is the ultimate goal. However, due to their unique features, biologics represent a specific challenge in formulation development. Most often, the main strategies used in the product development of small molecular weight drugs cannot be readily transferred into the product development of biologics. The modified/ improved strategies need to be applied to face the specific challenges linked to protein and peptide drugs. In addition, specific challenges and opportunities in nonclinical safety testing of biologics need to be addressed and optimized
II. CHALLENGES IN PRODUCT DEVELOPMENT FOR BIOLOGICS
A. Challenges Related to Route of Administration
Most biologic products available today are designed for parenteral (injected) administration, with many—like monoclonal antibody drugs—requiring intravenous infusions in clinical settings. Although some biologics are formulated for self-administration as subcutaneous (under the skin) injections, alternative methods of delivery could offer significant benefits. For example, switching from subcutaneous injections to another method could improve patient comfort, compliance, and dosing accuracy for treatments like human growth hormone, where precision in dosing is essential but hard to achieve with current methods.
B. Formulation Properties for Parenteral Administration
The parenteral route also presents challenges in formulation. For instance, as the concentration of biologics increases, so does the viscosity, making it difficult to inject. Furthermore, factors like pH and osmolality need to be optimized to ensure patient safety and comfort. Advances in nanotechnology and the development of delivery systems, such as micro- and nanoparticles, have created new options. Specially designed lipid or polymer-based carriers can help deliver these complex drugs. However, certain formulations require larger particles, which then need larger-gauge needles, limiting convenience and ease of use.
C. Optimizing the Dosage Regimen
Biologics typically need to be administered in high doses and at frequent intervals because they are poorly absorbed through the gastrointestinal tract. Although strategies like mucoadhesive patches and cell-penetration peptides are being developed for oral delivery, more research is needed to confirm their long-term safety. Sustained-release formulations, which allow for less frequent administration and greater safety, are promising but require significant changes in the molecule, which can be costly and require new regulatory approval. To improve convenience, companies are focusing on encapsulation technologies for slow-release formulations, though these are also more costly to manufacture.
D. Overcoming Absorption and Metabolism Barriers
The development of biologics involves complex challenges related to absorption, distribution, metabolism, and elimination (ADME). Unlike small molecules, biologics face species-specific variations in how they are processed, which complicates the transfer of data from animal studies to human applications. The processes by which biologics are cleared by the liver and the immune system, along with potential toxicity, remain areas that need more exploration.
III. ROUTES OF ADMINISTRATION FOR BIOLOGICS
A. Parenteral Route
Most biologics are currently administered via parenteral routes, typically through intravenous or subcutaneous injections. Although effective, this method poses challenges such as limited control over drug distribution and metabolism, as well as an invasive delivery method. Additionally, issues related to the biologics’ absorption, distribution, metabolism, and elimination (ADME) can be unpredictable, creating hurdles in consistent dosing and therapeutic outcomes.
B. Oral Route
The oral route is considered ideal for chronic therapies due to its ease of administration and patient compliance. However, the gastrointestinal (GI) tract poses significant challenges for biologics, as these large molecules are susceptible to harsh acidic conditions and digestive enzymes, leading to low bioavailability. Attempts to improve oral bioavailability include modifying the biologic molecule itself (e.g., using PEGylation to protect against GI enzymes) and encapsulating biologics in nanoparticles or liposomes for better protection. While these approaches have shown promise in laboratory settings, they have yet to achieve significant success in clinical applications due to issues with enzyme stability and bioavailability.
C. Intranasal Route
The intranasal route provides a noninvasive alternative, offering rapid absorption and bypassing first-pass metabolism. This method has potential for small molecule drugs and may be beneficial for certain biologics, particularly for targeting central nervous system disorders like Alzheimer’s or Parkinson’s disease, as it can access the brain via the olfactory pathway.
However, challenges such as short residence time, enzymatic barriers, and low mucosal absorption limit its effectiveness for biologics. Technologies such as chitosan-based carriers and mucoadhesive microspheres are being explored to improve absorption and retention.
D. Pulmonary Route
The pulmonary route is another noninvasive option, beneficial for conditions requiring quick drug action and systemic delivery. Although inhalable biologics avoid the harsh GI environment, the respiratory tract presents its own challenges, such as the mucus barrier and variable absorption depending on particle size and lung physiology. Despite setbacks, such as the limited success of inhaled insulin, research in aerosol delivery systems continues, with the aim of achieving effective, stable delivery.
E. Transdermal Route
The transdermal route involves delivering biologics through the skin, typically via microneedles or patches. This method offers a sustained-release mechanism that is easy for patients to self-administer. Microneedle patches for insulin and other biologics have shown promising results in animal studies, yet challenges remain regarding consistent penetration and metabolic stability. Ongoing studies focus on optimizing the patch materials and formulations to ensure efficient, controlled delivery.
IV. NEW DELIVERY DEVICES FOR BIOLOGICS
To address the high rate of treatment discontinuation caused by noncompliance, several innovative delivery devices have been developed, including prefilled syringes, manual injector pens, autoinjectors, and needle-free systems. These devices are designed to make administration simpler and less intimidating for patients, thereby improving adherence. In particular, micro- and nanomechanical devices have shown promise for precisely controlling drug release and delivery, allowing for optimized size and delivery properties.
These advancements are especially beneficial for pediatric, elderly, and arthritic patients, who often face difficulties with traditional needle-based methods. The goal is to create devices that meet the unique needs of these special populations, making treatment more accessible and manageable. Despite promising developments, more research and refinement are necessary to fully explore and expand these technologies to make biologic treatments more patient-friendly and improve bioavailability.
Biologics are becoming a significant area of focus in pharmaceutical development due to their potential to treat complex conditions. While progress has been made in manufacturing biologics, the development of efficient delivery systems remains challenging, with most still requiring invasive administration. Emerging technologies and innovative delivery approaches hold promise for enhancing the bioavailability and accessibility of biologics, paving the way for more advanced treatment options in the future.
[1] Kling J. Fresh from the biotech pipeline – 2011. Nat Biotechnol. 2012;30(2):128-131. [2] Nelson AL, Dhimolea E, Reichert JM. Development trends for human monoclonal antibody therapeutics. Nat Rev Drug Discov. 2010;9(10):767-774. [3] Ezan E. Pharmacokinetic studies of protein drugs: past, present and future. Adv Drug Deliv Rev. 2013;65(8):1065-1073. [4] Poelstra K, Beljaars L, Melgert BN. Cell-specific delivery of biologicals: problems, pitfalls and possibilities of antifibrotic compounds in the liver. Drug Discov Today. 2013;18(23-24):1237-1242. [5] Chung SW, Hil-lal TA, Byun Y. Strategies for non-invasive delivery of biologics. J Drug Target. 2012;20(6):481-501. [6] Leader B, Baca QJ, Golan DE. Protein therapeutics: a summary and pharmacological classification. Nat Rev Drug Discov. 2008;7(1):21-39. [7] Xu X, Vugmeyster Y. Challenges and opportunities in absorption, distribution, metabolism and excretion studies of therapeutic biologicals. AAPS J. 2012;14(4):781-791. [8] Martin-Moe S, Lim FJ, Wong RL, Sreedhara A, Sundaram J, Sane SU. A new roadmap for biopharmaceutical drug product development: integrating development, validation and quality by design. J Pharm Sci. 2011;100(8):3031-3043. [9] Richard J. Parenteral biologics delivery: recent progresses, key challenges and perspectives. Eur J Parenteral Pharm Sci. 2012;17(3):94-109. [10] Duncan R, Gaspar R. Nanomedicine(s) under the microscope. Mol Pharm. 2011;8(6):2101-2141. [11] Burnett JC, Rossi JJ. RNA-based therapeutics: current progress and future prospects. Chem Biol. 2012;19(1):60-71. [12] Kronenberg S, Baumann A, de Haan L, et al. Current challenges and opportunities in nonclinical safety testing of biologics. Drug Discov Today. 2013;18(23-24):1138-1143. [13] Burgess BE. Optimizing drug delivery for modern biologics. BioPharm Int. 2012;25(5):30-32. [14] Cázares-Delgadillo J, Ganem-Rondero A, Kalia YN. Human growth hormone: new delivery systems, alternative routes of administration, and their pharmacological relevance. Eur J Pharm Biopharm. 2011;78(2):278-288. [15] Reithmeier H, Hermann J, Göpferich A. Lipid microparticles as a parenteral controlled release device for peptides. J Control Release. 2001;73(2-3):339-350. [16] Govardhan C, Khalaf N, Jung CW, et al. Novel long-acting formulation of human growth hormone. Pharm Res. 2005;22(9):1461-1470. [17] Whitehead K, Shen Z, Mitragotri S. Oral delivery of macromolecules using intestinal patches: applications for insulin delivery. J Control Release. 2004;98(1):37-45. [18] Khafagy el-S, Morishita M. Oral biodrug delivery using cell-penetrating peptide. Adv Drug Deliv Rev. 2012;64(6):531-539. [19] Shi J, Votruba AR, Farokhzad OC, Langer R. Nanotechnology in drug delivery and tissue engineering: from discovery to applications. Nano Lett. 2010;10(9):3223-3230. [20] Sonaje K, Lin YH, Juang JH, Wey SP, Chen CT, Sung HW. In vivo evaluation of safety and efficacy of self-assembled nanoparticles for oral insulin delivery. Biomaterials. 2009;30(12):2329-2339. [21] Krol S, Ellis-Behke R, Marchetti P. Nanomedicine for treatment of diabetes in an aging population: state-of-the-art and future developments. Nanomedicine. 2012;8(Suppl 1):S69-S76. [22] Cone RA. Barrier properties of mucus. Adv Drug Deliv Rev. 2009;61(2):75-85.
Copyright © 2024 Shrikant Subhash Garud , Samadhan Ananda Dhatrak , Thube Rushikesh Sayaji, Amit Ambadas Chaudhari. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Paper Id : IJRASET65057
Publish Date : 2024-11-07
ISSN : 2321-9653
Publisher Name : IJRASET
DOI Link : Click Here